WO2001083674A1 - Biological identification system with integrated sensor chip - Google Patents

Biological identification system with integrated sensor chip Download PDF

Info

Publication number
WO2001083674A1
WO2001083674A1 PCT/US2001/014257 US0114257W WO0183674A1 WO 2001083674 A1 WO2001083674 A1 WO 2001083674A1 US 0114257 W US0114257 W US 0114257W WO 0183674 A1 WO0183674 A1 WO 0183674A1
Authority
WO
WIPO (PCT)
Prior art keywords
reagent
biosensor
electrodes
electrode
electrically conductive
Prior art date
Application number
PCT/US2001/014257
Other languages
French (fr)
Inventor
Jen-Jr Gau
Original Assignee
Gau Jen Jr
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gau Jen Jr filed Critical Gau Jen Jr
Priority to CA2407973A priority Critical patent/CA2407973C/en
Priority to AU6114501A priority patent/AU6114501A/en
Priority to JP2001580284A priority patent/JP4949589B2/en
Priority to AU2001261145A priority patent/AU2001261145B2/en
Priority to EP01935016A priority patent/EP1278821A4/en
Publication of WO2001083674A1 publication Critical patent/WO2001083674A1/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/508Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above
    • B01L3/5088Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above confining liquids at a location by surface tension, e.g. virtual wells on plates, wires
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N27/00Investigating or analysing materials by the use of electric, electrochemical, or magnetic means
    • G01N27/26Investigating or analysing materials by the use of electric, electrochemical, or magnetic means by investigating electrochemical variables; by using electrolysis or electrophoresis
    • G01N27/28Electrolytic cell components
    • G01N27/30Electrodes, e.g. test electrodes; Half-cells
    • G01N27/327Biochemical electrodes, e.g. electrical or mechanical details for in vitro measurements
    • G01N27/3275Sensing specific biomolecules, e.g. nucleic acid strands, based on an electrode surface reaction
    • G01N27/3277Sensing specific biomolecules, e.g. nucleic acid strands, based on an electrode surface reaction being a redox reaction, e.g. detection by cyclic voltammetry
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • B01J2219/00317Microwell devices, i.e. having large numbers of wells
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00596Solid-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/0061The surface being organic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00612Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports the surface being inorganic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00617Delimitation of the attachment areas by chemical means
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00617Delimitation of the attachment areas by chemical means
    • B01J2219/00619Delimitation of the attachment areas by chemical means using hydrophilic or hydrophobic regions
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/0063Other, e.g. van der Waals forces, hydrogen bonding
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00653Making arrays on substantially continuous surfaces the compounds being bound to electrodes embedded in or on the solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/00702Processes involving means for analysing and characterising the products
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00722Nucleotides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00725Peptides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B81MICROSTRUCTURAL TECHNOLOGY
    • B81CPROCESSES OR APPARATUS SPECIALLY ADAPTED FOR THE MANUFACTURE OR TREATMENT OF MICROSTRUCTURAL DEVICES OR SYSTEMS
    • B81C2201/00Manufacture or treatment of microstructural devices or systems
    • B81C2201/01Manufacture or treatment of microstructural devices or systems in or on a substrate
    • B81C2201/0101Shaping material; Structuring the bulk substrate or layers on the substrate; Film patterning
    • B81C2201/0128Processes for removing material
    • B81C2201/013Etching
    • B81C2201/0133Wet etching
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/10Libraries containing peptides or polypeptides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/14Apparatus specially adapted for use in combinatorial chemistry or with libraries for creating libraries

Definitions

  • the field of the present invention relates generally to apparatus and methods for sensing or detecting various target analytes, including ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein) and in particular, to biosensors, methods of using the biosensors, and methods for making the biosensors.
  • ionic molecules e.g. iron, chromium, lead, copper, calcium or potassium
  • macromolecules e.g. DNA, RNA or protein
  • a variety of biosensors have been developed for the detection of biological material, such as pathogenic bacteria.
  • Conventional methods for detecting bacteria usually involve a morphological evaluation of the organisms and rely on (or often require) growing the number of organisms needed for such an evaluation; such methods are time consuming and are typically impractical under field conditions.
  • the need for rapid detection as well as portability has led to the development of systems that couple pathogen recognition with signal transduction.
  • Both optical and electrochemical detection of bacteria have been reported (Ivnitski, et. al., 1999, T. Wang, et. al., 2000). (Full citations to the literature cited herein are given at the end of the specification.)
  • the present inventive entity has determined that electrochemical methods have an advantage in that they are more amenable to miniaturization.
  • Requirements for an ideal detector include high specificity and high sensitivity using a protocol that can be completed in a relatively short time. Moreover, systems that can be miniaturized and automated offer a significant advantage over current technology, especially if detection is needed in the field.
  • the electrochemical methods use the principle of electrical circuit completion.
  • a counter electrode is used to provide a return path to the sample solution or reagent and a reference electrode is used as a reference point against which the potential of another electrode or electrodes are determined (typically that of the working electrode or measuring electrode). Since this contact must be provided by electrochemical means, i.e. a metal electrode immersed in a chemical solution or reagent, it is impossible to avoid generating an electrical potential in series with the potential developed by the electrode.
  • the conventional theory in the electrochemical methods is that it is essential for the reference electrode potential to be very stable and not be affected by chemical changes in the solution.
  • silver/silver chloride reference electrodes which provide a very stable reference potential, are the most common type of electrode used for reference electrodes today.
  • the typical silver/silver chloride reference electrode 10 contains a chloridised silver wire 1 (a layer of silver chloride coated on silver wire) immersed in a solution 5 of potassium chloride (3.5M KC1) saturated with silver chloride (AgCl).
  • This internal filling solution 5 slowly seeps out of the electrode 10 through a porous ceramic junction 20 and acts as an electrical connection between the reference element 1 and the sample.
  • Potassium chloride is used because it is inexpensive and does not normally interfere with the measurement.
  • the solution 5 also includes silver chloride to prevent dissolution of the coating on the reference element 1. It is therefore necessary to maintain the level of solution 5 in the electrode 10 using a filling solution hole 40.
  • the present inventive entity has determined that robust and precise biological detection can be achieved using electrochemical methods, and more precisely using redox (reduction-oxidation) methods, without the need of a two-layered reference electrode (e.g., silver chloride coated on silver) having a known reference electrode potential described above.
  • redox reduction-oxidation
  • potentiostat 50 which is a control amplifier with the test cell placed in the feedback loop.
  • the objective is to control the potential difference between a test electrode (working electrode) 60 and a reference electrode 70 by the application of a current via the third, auxiliary electrode (counter electrode) 80.
  • a fairly good potentiostat may be built using a minimum of components.
  • 90A and 90B are 1.22V bandgap reference diodes connected between the positive and negative power rails. Potentiometer 100 is then used to set the required cell polarization, applied to the non-inverting input of the main amplifier 110.
  • the working electrode 60 connects to the ground of the circuit and the reference electrode 70 to the inverting input of the main amplifier 110.
  • a unity gain buffer amplifier 120 is used.
  • gain buffer amplifier 120 has a much higher bandwidth than amplifier 110, otherwise the circuit is likely to oscillate when driving a capacitative cell.
  • the output of the buffer amplifier 120 connects to the auxiliary electrode 80 via a current measuring resistor 140. Differential amplifier 130 is then used to measure the voltage drop across this resistor 140 and to convert it to a ground referenced output voltage.
  • Microelectromechanical systems (MEMS) technology provides transducers to perform sensing and actuation in various engineering applications.
  • MEMS technology is that it makes possible mechanical parts of micron size that can be integrated with electronics and batch fabricated in large quantities.
  • MEMS devices are fabricated through the process of micromachining, a batch production process employing lithography. Micromachining relies heavily on the use of lithographic methods to create 3-dimensional structures using pre-designed resist patterns (masks) (Ho and Tai, 1996, 1998).
  • MEMS is one suitable technology for making micro fabricated devices or aspects thereof.
  • Microfabricated devices are generally defined as devices fabricated by using MEMS and/or integrated circuit (IC) technology.
  • An integrated circuit (IC) is defined as a tiny chip of substrate material upon which is etched or imprinted a complex of electronic components and their interconnections.
  • the present inventive entity has determined that miniaturization and portability features are inherent in electrochemical methods make these methods excellent candidates for integration into MEMS devices, and, further that it would be advantageous to integrate MEMS technology with biosensing methods to detect various ionic molecules and macromolecules (DNA, RNA or protein).
  • the present inventive entity has also determined that in the area of biosensing of bacteria, one of the most effective means of achieving high specificity is to detect the bacteria's genetic material (e.g. rRNA, mRNA, denatured DNA).
  • rRNA e.g. rRNA, mRNA, denatured DNA
  • ssDNA single- stranded DNA
  • monitoring the hybridization event allows selective sensing of target cells.
  • coupling the hybridization event with an enzymatic reaction leads to signal amplification, as each substrate-to-product turnover contributes to the overall signal.
  • Biosensing to detect DNA hybridizations that are amplified by enzymatic reaction can still be completed within a reasonably short time according to the invention.
  • E. coli Escherichia coli
  • MEMS microelectrometric microsenors
  • SAMs self-assembled monolayers
  • DNA hybridization DNA hybridization
  • enzyme amplification all contribute to the design of a miniaturized, specific, and sensitive E. coli detector.
  • DNA electrochemical probes have been reported previously (Wang, et al., 1997, Marrazza, et al., 1999), with graphite or carbon electrodes typically used.
  • Commercial units for amperometric detection of DNA from E. coli using screen-printed carbon electrodes on disposable test strips are also available.
  • Screen- printing has the advantage of low cost; however, achieving high dimensional precision is not easy.
  • the present inventive entity has determined a need to develop a method of using lithography to accurately pattern in ⁇ m size dimensions a wide range of materials such as metals (e.g. Au, Ag) and carbon.
  • a surface modification such as self-assembly monolayer (SAM) of biotin-DAD-C12-SH dodecanamide, is a preferred method of selectively immobilizing molecules on MEMS surfaces.
  • SAM self-assembly monolayer
  • the present invention identifies and takes advantage of certain benefits inherent in each technology. Using DNA hybridization and enzyme amplification, the present invention achieves the required specificity and sensitivity. Using MEMS and SAMs, the present inventive entity fabricated a miniaturized system that can be developed into a portable instrument. Finally, the invention demonstrates that the present detection system is applicable to a broad range of pathogenic bacteria. For example, the detection module and assay protocol can be adapted to detect uropathogenic E. coli and identify microorganisms causing otitis media (middle ear infection).
  • a first aspect of the present invention provides an apparatus and method for sensing or detecting various target analytes, including especially macromolecules (e.g.
  • the substrated biosensor system comprises at least two electrodes that are typically fabricated together as a single series microfabrication process step on the substrate. However, successive microfabrication steps may also be employed to fabricate the system.
  • the electrodes are all made out of pure metal (as opposed to the Ag/AgCl electrodes of the prior art, for exmple).
  • the substrated biosensor system comprises a working electrode, a reference electrode and a counter (auxiliary) electrode.
  • the substrated biosensor system has the same electrochemical performance as a conventional electrochemical biosensor.
  • the substrated biosensor system should be compatible with integrated circuit (IC) and/or MEMS fabrication processes and be capable of being constructed in a small area.
  • a second aspect of the present invention provides an apparatus and method for confinement of reagent and/or solution in a biosensor using surface tension at small scale.
  • the reagent and/or solution contain the necessary electrolytes and/or analytes needed for biological sensing.
  • the apparatus and method in the present invention allow for each electrode used for sensing or detecting various ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein) to be in selective contact with the reagent and/or solution when the electrolytes and/or analytes are needed by using controllable surface properties and surface tension forces at small scale.
  • various ionic molecules e.g. iron, chromium, lead, copper, calcium or potassium
  • macromolecules e.g. DNA, RNA or protein
  • the apparatus and method for confinement of reagent and/or solution (and thus the electrolytes and/or analytes in the reagent and/or solution) using surface tension is incorporated with a portable or handheld device and is immune to shaking of the device.
  • the reagent and/or solution should be held firmly in position by the biosensor using surface tension even when the biosensor is flipped upside down.
  • a third aspect of the present invention provides an apparatus and method for integrating the components of an electrochemical sensor and/or sensors (e.g. electrodes) and additional required electronic circuit components (e.g. amplifiers) in an electrochemical sensor or sensors with integrated circuit (IC) technologies.
  • the entire sensor system and/or systems can be incorporated on a single IC substrate or chip, such as a single semiconductor (e.g. silicon or gallium arsenide) substrate or chip.
  • a single semiconductor e.g. silicon or gallium arsenide
  • no or much fewer external components and/or instruments are required to complete the system or systems.
  • the sensor and/or sensors are preferably fabricated using the IC process.
  • a preferred feature of the invention is to modify the surface on at least one of the electrodes.
  • the surface is modified for anchoring macromolecules on the surface.
  • the surface is modified using a self-assembly monolayer (SAM) such as biotin- streptavidin.
  • SAM self-assembly monolayer
  • the SAM is placed on the surface of a working electrode in an electrochemical sensor.
  • materials such as sol gel and/or carbon paste may be used to modify the surface (as a replacement for SAM or in combination with SAM).
  • Fig. 1 is a schematic of an embodiment of a conventional reference electrode.
  • Fig. 2 is a schematic of an embodiment of a potentiostat used in the electrochemical methods.
  • Fig. 3 is a schematic of an embodiment of a plurality of electrochemical biosensors on a circular substrate wafer according to the present invention.
  • Fig. 4 is a schematic of alternative embodiment of a plurality of electrochemical biosensors on a square substrate according to the present invention.
  • Fig. 5 is a schematic of another embodiment of an electrochemical biosensor on a substrate according to the present invention.
  • Fig. 6 is a graph of Cyclic Voltammetry (CV) scan potential over time.
  • Fig. 7 is a one-cycle Cyclic Voltammetry (current vs. bias potential) taken by a biosensor according to the present invention.
  • Fig. 8. is a Cyclic Voltammetry (current vs. bias potential) at different scan rate taken by a biosensor according to the present invention.
  • Fig. 9. is a graph of the square root of scan rate vs. peak current taken by a biosensor according to the present invention.
  • Fig. 10 is a Cyclic Voltammetry (current vs. bias potential) on a plurality of cycles at constant scan rate taken by a biosensor according to the present invention.
  • Figs 11(a), (b) and (c) are diagrams showing reagent and/or solution confinement by surface tension and treatment according to the present invention.
  • Fig. 12 is a diagram showing reagent and/or solution confinement selectively over only a working electrode on a biosensor according to the present invention.
  • Fig. 13 is a diagram showing reagent and/or solution confinement over all electrodes on a biosensor according to the present invention.
  • Fig. 14 is a side view diagram showing the first step of how an embodiment of a biosensor of the present invention can be fabricated.
  • Fig. 15 is a diagram showing the second step of how the biosensor can be fabricated.
  • Fig. 16 is a diagram showing the third step of how the biosensor can be fabricated.
  • Fig. 17 is a diagram showing the fourth step of how the biosensor can be fabricated.
  • Fig. 18 is a diagram showing the fifth step of how the biosensor can be fabricated.
  • Fig. 19 is a diagram showing the sixth step of how the biosensor can be fabricated.
  • Fig. 20 is a diagram showing how the surface of an embodiment of the present biosensor can be modified to prevent non-specific binding.
  • Fig. 21 is a diagram showing the final result on the biosensor surface of the surface modification process in Fig. 20.
  • Fig. 22 is a sectional diagram of an embodiment of the present invention of a biosensor system with a biosensor integrated with integrated circuit components.
  • Fig. 23 is a diagram of an embodiment of the present invention of a plurality of biosensors integrated with integrated circuit components.
  • Fig. 24 is a sectional diagram of a biosensor unit that, together with other similar units, makes up the plurality of biosensors integrated with the integrated circuit components in Fig. 24.
  • Fig. 25 is a diagram showing the first step of how an embodiment of a biosensor (sensor-chip) of the present invention can be fabricated by integrated circuit (IC) technology with a CMOS device integrated on the biosensor (sensor-chip) itself.
  • IC integrated circuit
  • Fig. 26 is a diagram showing the second step of how the biosensor (sensor-chip) can be fabricated by integrated circuit (IC) technology.
  • Fig. 27 is a diagram showing the third step of how the biosensor (sensor-chip) can be fabricated by IC technology.
  • Fig. 28 is a diagram showing the fourth step of how the biosensor (sensor-chip) can be fabricated by IC technology.
  • Fig. 29 is a diagram showing the fifth step of how the biosensor (sensor-chip) can be fabricated by IC technology.
  • Fig. 30 is a diagram showing the sixth step of how the biosensor (sensor-chip) can be fabricated by IC technology.
  • Fig. 31 is a diagram showing how an embodiment of a biosensor of the present invention can be use to detect ionic analytes (or molecules).
  • Fig. 32 is Cyclic Voltammetry (CV) (current vs. bias potential) graph taken by the sensor of Fig. 31.
  • Fig. 33 is a diagram showing the first step of how an embodiment of a biosensor of the present invention can be used to detect macromolecules (e.g. DNS, RNA, protein).
  • macromolecules e.g. DNS, RNA, protein
  • Fig. 34 is a diagram showing the second step of the detection of the macromolecules.
  • Fig. 35 is a diagram showing the third step of the detection of the macromolecules.
  • the first aspect of the invention relates to detection of various target analytes, especially ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein), using the principles of electrochemical detection.
  • ionic molecules e.g. iron, chromium, lead, copper, calcium or potassium
  • macromolecules e.g. DNA, RNA or protein
  • the redox cell is a device that converts chemical energy into electrical energy or vice versa when a chemical reaction occurs in the cell.
  • the cell consists of three electrodes immersed into an aqueous solution (electrolyte) with electrode reactions occurring at the electrode-solution surfaces.
  • the cell consists of two electronically conducting phases (e.g., solid or liquid metals, semiconductors, etc.) connected by an ionically conducting phase (e.g. aqueous or nonaqueous solution, molten salt, ionically conducting solid).
  • an electrical current passes, it must change mode from electronic current to ionic current and back to electronic current.
  • ionically conducting phase e.g. aqueous or nonaqueous solution, molten salt, ionically conducting solid.
  • Each electrochemical reaction is reduction-oxidation (redox) reaction that occurs in the redox cell.
  • redox reduction-oxidation
  • the hydrogen for example, is oxidized at the anode electrode by transferring electrons to the anode electrode and the oxygen is reduced at the cathode electrode by accepting electrons from the cathode electrode.
  • the overall electrochemical reaction is the sum of the two electrode reactions.
  • the ions produced in the electrode reactions in this case positive hydrogen ions and negative hydroxyl ions, will recombine in the solution to form the final product of the reaction: water.
  • the electrons are conducted from the anode electrode to the cathode electrode through an outside electric circuit where the electronic current can be measured.
  • the reaction can also be reversed; water can be decomposed into hydrogen and oxygen by the application of electrical power in an electrolytic cell.
  • a three-electrode system of the invention is an electrochemical cell containing a working electrode, a counter electrode (or auxiliary electrode), and a reference electrode.
  • a current may flow between the working and counter electrodes, while the potential of the working electrode is measured against the reference electrode.
  • This setup can be used in basic research to investigate the kinetics and mechanism of the electrode reaction occurring on the working electrode surface, or in electroanalytical applications.
  • the detection module in the preferred embodiment in the instant invention is based of a three- electrode system.
  • the counter electrode is used to make an electrical connection to the electrolyte so that a current can be applied to the working electrode.
  • the counter electrode is usually made of inert materials (noble metals or carbon/graphite) to avoid its dissolution. It has been observed in connection with the present invention that a small feature or small cross- section at the counter electrode will heat up the surrounding solution when a large current is pulled out from the counter electrode. Bubbles will be generated if the current is continuously overflowed, and ultimately dissolution of the electrode occurs. Thus, the bubble formation can be avoided by controlling the current and/or the electrode size. In a preferred embodiment of the present invention, the width of the counter electrode is large enough to avoid this heat up problem even at large current.
  • the reference electrode is used as a reference point against which the potential of other electrodes (typically that of the working electrode or measuring electrode) can be measured in an electrochemical cell.
  • the few commonly used (and usually commercially available) electrode assemblies all have an electrode potential independent of the electrolyte used in the cell, such as a silver/silver-chloride electrode, calomel electrode, or hydrogen electrode.
  • the present inventive entity has determined that a single layer electrode, such as a single layer of gold electrode, can fulfill the requirement for a reference electrode.
  • other materials such as silver, copper, platinum, chromium, aluminum, titanium, nickel may also work as a single layer reference electrode under the right conditions.
  • the working electrode plays a central role in the electrochemical biosensors of the invention.
  • the reaction occurring at the working electrode may be used to perform an electrochemical analysis of the electrolyte solution. It can serve either as an anode or a cathode, depending on the applied polarity.
  • One of the electrodes in some "classical two- electrode” cells can also be considered a “working" ("measuring,” “indicator,” or “sensing") electrode, e.g., in a potentiometric electroanalytical setup where the potential of the measuring electrode (against a reference electrode) is a measure of the concentration of a species in the solution.
  • the counter and reference electrodes are configured to extend generally about the periphering of the working electrode of the biosensor. Suitability configurations are seen in, for example, Figs. 3, 4, 5, 12, 13 and 23.
  • the electrochemical biosensor is fabricated using microelectromechanical systems (MEMS) technology. Referring now to Fig. 3, a layer of silicon dioxide (SiO 2 , lOOOA) is deposited on a bare silicon wafer 200 (prime grade, p- type ⁇ 100>, thickness 500-550. ⁇ m) and served as a pad layer underneath the silicon nitride (Si 3 N , lOOOA) to release stress and improve adhesion.
  • a plurality of MEMS biosensors (such as biosensor 210) were fabricated with working electrodes of various dimensions (such as working electrode 220). Preferably each of the working electrodes is etched to form a well up to 350 ⁇ m in depth.
  • the nitride-coated silicon wafer 200 was patterned and bulk etched using KOH along the [111] and [100] crystal planes, and the depth of the well was controlled by KOH etching time and temperature.
  • the 100. ⁇ m wide auxiliary (such as auxiliary electrode 230) and reference (such as reference electrode 240) electrodes are separated from their corresponding working electrode 220 by 200 ⁇ m.
  • Fig. 3 shows a schematic of the pattern used in generating the MEMS biosensors (such as biosensor 210).
  • nitride and oxide were removed by HF etching to release internal stress, and another oxide layer (5000A) was deposited for electrical isolation. Electrodes were patterned by PR5214 photo resist reverse imaging and lift-off process with electron beam deposition of Au(200 ⁇ A)/Cr(20 ⁇ A). Finally the wafer 200 was bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding Si areas.
  • HMDS hexamethyldisilazane
  • the hydrophobic nature of the surrounding area along with the 3 -dimensional nature of the working electrode (such as working electrode 220), allows containment of a liquid droplet on the working electrode 220 during the initial sensing step of immobilizing to the working electrode molecules of interest in the solution.
  • This design effectively minimized nonspecific binding of biomolecules to other areas of the MEMS array.
  • the material used for all the electrochemical electrodes is gold (Au).
  • Fig. 4 shows an embodiment of biosensors fabricated on a square substrate 300 (such as silicon, gallium arsenide, plastic and/or glass) having a plurality of circular biosensors, such as biosensor 310.
  • the biosensor 310 comprises a working electrode 320, a reference electrode 330, and a counter (auxiliary) electrode 340.
  • the electrodes are constructed out of a single layer of conducting materials, available in MEMS technology.
  • all of the electrodes should preferably be constructed out of gold (Au).
  • Fig. 5 shows yet another preferred embodiment of the present invention.
  • the biosensor 410 comprises a working electrode 420, a reference electrode 430 and a counter (auxiliary) electrode 440 fabricated on the substrate.
  • the electrodes are made out of gold.
  • the working electrode 420 is formed in a built-in well 450 in the substrate up to 350 ⁇ m in depth.
  • the well 450 is designed for confining a desired reagent within the well-defined space.
  • the well 450 fabricated by the microfabrication methods described herein is bordered by (111) silicon planes after KOH etching.
  • the working electrode 420 defined by the microfabrication methods described above, covers the entire well 450 surface.
  • the above embodiments have the advantage of being easy to fabricate and can be fabricated together as a single series microfabrication process step on the substrate.
  • successive microfabrication steps may also be employed for fabrication.
  • these low-cost and easy-to-fabricate biosensors are reusable and have the same robust and reversible electrochemical performance as conventional biosensors.
  • Cyclic Voltammetry analytical technique is used.
  • CV is one of the most versatile analytical techniques used in the study of electroactive species and the characterization of biosensors. It is widely used as both an industrial and academic research tool in the fundamental characterization of electrochemical systems.
  • E 0 initial potential
  • E m maximum potential
  • V/sec sweep rate
  • Fig. 6 illustrates this concept. Repeated cycles of reduction and oxidation of the analyte generate alternating anodic and cathodic currents in and out of the working electrode. Since the solution and/or reagent is not stirred, diffusion effects are observed at different analyte concentrations and different scan rates.
  • the potential is graphed along the x-axis with more positive (or oxidizing) potentials plotted to the right, and more negative (or reducing) potentials to the left.
  • the current is plotted on the y-axis with cathodic (i.e., reducing) currents plotted down along the negative direction, and anodic (i.e. oxidizing) currents plotted in the positive direction.
  • the analyte used in the following control experiment was potassium ferricyanide, K 3 Fe(CN) 6 (329.26 g/mol), which contains an iron atom in the +3 oxidation state (Fe ⁇ ) in a buffer solution of potassium nitrate, KNO (101.11 g/mol).
  • KNO potassium nitrate
  • a single electron can be added to the ferricyanide anion. This will cause it to be reduced to the ferricyanide anion, Fe"(CN) 6 4' , which contains an iron atom in the +2 oxidation state (Fe 11 ).
  • Fe 11 iron atom in the +2 oxidation state
  • This simple, one electron exchange between the analyte and the electrode is a well behaved, reversible reaction. This means that the analyte can be easily reduced to Fe ⁇ (CN) 6 4" and then easily oxidized back to Fe ⁇ (CN) 6 3" .
  • a redox couple is a pair of analytes differing only in oxidation state.
  • the voltammogram shown in Fig. 8 exhibits two asymmetric peaks, one cathodic
  • ip C the other anodic (i pa ).
  • a standard reference electrode such as the normal hydrogen electrode (NHE)
  • the formal potential associated with this half-reaction is near +358 mV.
  • the working electrode is held at a potential more positive than +400 mV, then the analyte tends to be oxidized to the Fe'"(CN) 6 3 ⁇ form. This oxidation at the working electrode causes electrons to go into the electrode from the solution resulting in an anodic current.
  • potentials more negative than +400 mV the analyte tends to be reduced to Fe'" (CN) 6 ⁇ . This reduction at the working electrode causes electrons to flow out of the electrode into the solution resulting in a cathodic current.
  • the preferred sensor design embodiment of the present invention does not utilize a standard reference electrode like NHE, silver/silver chloride (Ag/AgCl) or saturated calomel electrode (SCE), and since all three electrodes are gold (Au), the rest (unbiased) potential in this experiment is close to zero volts.
  • a standard reference electrode like NHE, silver/silver chloride (Ag/AgCl) or saturated calomel electrode (SCE)
  • Au gold
  • the important parameters of a cyclic voltammogram are the magnitudes of the cathodic and anodic peak currents (i pc and i pa , respectively) and the potentials at which these currents are observed (E pc and E pa , respectively). Using these parameters, it is possible to calculate the formal reduction potential (E 0 - which is centered between E pa and E pc ) and the number of electrons (n) transferred in the charge transfer reaction.
  • the Randles-Sevcik equation predicts that the peak current should be proportional to the square root of the sweep rate when voltammograms are taken at different scan rates. As shown in Fig. 9, the plot of peak current versus the square root of sweep rate yields a straight line.
  • the Randles-Sevcik equation can be modified to give an expression for the slope of this straight line as follows,
  • the scan rate dependence of the peak potentials and peak currents are used to evaluate the number of electrons participating in the redox reaction as well as provide a qualitative account of the degree of reversibility in the overall reaction.
  • Fig. 10 shows the CV scan for 12 consecutive cycles with minor deviations from the first cycle.
  • a second aspect of the present invention relates to confinement of a reagent and/or solution in a biosensor using surface tension at small scale.
  • the reagent and/or solution contains the target analyte(s) and/or the chemical(s) needed for biosensing.
  • a biosensor's performance is mainly determined by its specificity and sensitivity.
  • the concept of confining the reagent on a substrate, such as silicon, was discovered in investigations relating to the present invention to be a solution for reducing the high level of detection noise caused by the non-specific binding of the analyte or other reagent or solution components, such as Horseradish Peroxidase (HRP), onto regions arund the periphery of the working electrode and causing high detection noises and creating high false positive rates.
  • HRP is used in the instant embodiment as a signaling enzyme. To verify that noise does come from the HRP residual at the surface, a simple test was done to estimate the contribution of this unwanted binding. HRP was introduced to a bare silicon chip, followed with several wash steps before the addition of the substrate solution. A very high level of enzymatic reaction was observed immediately after adding the substrate solution.
  • HRP like other proteins, sticks to the silicon surface easily and tightly.
  • Several commercial wash solutions and blocking protein were tested without any significant improvement of reducing non-specific binding.
  • the area outside the working electrode of the biosensor of the invention need not encounter the HRP solution in order to achieve biological sensing.
  • a reagent or solution containing components capable of non-specific binding may initially be confined to the well area to achieve the desired binding to the electrode in the well (preferably the working electrode), while avoiding non-specific binding to the periphery region of the biosensor.
  • the reagent or solution can then be washed off the biosensor and additional reagents and/or solutions later added to complete the sensing process.
  • the surface of the periphery area preferably should be protected by other mechanisms.
  • Silane-based molecules with various functional terminal groups have been used to modify surface properties of silicon wafers, silicon nitride chips, and atomic force microscopy (AFM) tips.
  • Silane compounds for the surface modification form robust monolayers chemically tethered to silicon oxide surfaces as a result of hydrolysis of terminal Si(Cl) n or SiO-C 2 H 5 groups.
  • the performance of artificial materials in contact with biological systems is determined by the surface interactions of the two materials. Since the surface interaction could result in noise increasing and structure damage, surface modification is one way to avoid this problem. The easiest way is to change the surface property of the structure or by using plastic.
  • the most widely implemented approach uses a self-assembly monolayer (SAM) from thiol molecules by chemisorption onto a gold surface and silane molecules to form SAM on a silicon oxide surface according to well- established procedures developed in the 80's mostly by C. D. Bain, and G. M. Whitesides.
  • SAM self-assembly monolayer
  • Other materials such as sol gel and carbon paste may also be used to modify the surface.
  • Fig. 11(a) the reagent is shaped by the hydrophilic working electrode and a droplet is nicely formed over the Au working electrode surface.
  • Fig. 11(b) demonstrates that with increasing volume of the reagent, the area covered by the reagent will gradually expand and then cover the other two electrodes.
  • Fig. 11(c) shows that when an excess stream of reagent is administered by pipette, a ball 465 of reagent will be formed instead of spreading out on the hydrophobic silicon substrate.
  • each individual electrode will be in contact with electrolyte and/or analyte and other components 509 only when needed by using controllable surface property and surface tension force at small scale.
  • all electrodes have to be immersed in the electrolyte and/or analyte 509 only at detection time. For most applications, the majority of time is spent on sample preparation as depicted in Fig. 12 to immobilize the electrolyte and or analyte 509 onto working electrode 510.
  • the coverage of the electrolyte and/or analyte 509 over the electrodes is controlled by surface property and reagent volume 520.
  • the above embodiments take advantage of surface tension at small scale to confine the electrolytes and or analytes and other components.
  • the above embodiments require much less reagent (in the order of pL to mL); and eliminate the need for bulk solution.
  • the above embodiments have the advantages of having the analyte close to the electrodes (pm to mm), whereby adequate exposure of analyte to the electrodes can be achieved using diffusion effects alone, and the need for stirring or other mixing is eliminated.
  • Another advantage of the above embodiments is the ease of the control and/or change of the coverage of electrolyte and/or analyte over the electrodes.
  • the above reagent and/or solution confinement embodiments reduce the loss of target analytes and improve the sensitivity of the biosensor by greatly reduce non-specific binding.
  • the above embodiments are relatively immune to shaking of the biosensor and are suitable for portable or handheld systems.
  • the reagent will be held firmly by surface tension even the sensor is flipped upside down.
  • the biosensor embodiments can be equipped with a reaction well 530 to help hold the reagent 520 in place and to control the shape of the reagent 520.
  • a reaction well 530 is not required and the confinement of the reagent 520 can be achieved by a simple flat surface.
  • Fig. 14 shows the first step of a preferred fabrication of an embodiment of the present biosensor (sensor-chip).
  • a layer of silicon dioxide (SiO 2 , lOOOA) 620 is first deposited on a bare Si wafer 610 (prime grade or test grade, p-type or n-type ⁇ 100>, thickness 500-550 ⁇ m).
  • the silicon dioxide 620 serves as a pad layer underneath the silicon nitride 630 (Si 3 N 4 , lOOOA) to release stress and improve adhesion.
  • the nitride wafer 600 is patterned (Fig. 15) by mask 640 and bulk etched using KOH along the [111] and [100] crystal planes (Fig. 16).
  • the etching creates a well 650 and the depth of the well 650 (up to 350 ⁇ m) is controlled by KOH etching time and temperature.
  • the nitride 630 and oxide 620 were then removed by HF etching to release internal stress, and another oxide 660 layer (5000A) is deposited for electrical isolation.
  • Figs. 18 and 19 show a lift-off process for the fabrication of the necessary electrodes.
  • the electrodes for biosensing are patterned on substrate 610 by using PR5214 photo resist layer 670 on silicon dioxide layer 660.
  • a mask 671 is used to transfer a desired pattern onto photo resist layer 670 by using reverse imaging process (which includes the removal unwanted photo resist layer 670).
  • a layer of gold Au (2000A) 680 is electron-beam deposited on silicon dioxide layer 660 with the desired photo resist pattern layer 670.
  • a deposition of an adhesive 690 such as chromium (Cr 20 ⁇ A) occurs before the deposition of Au(200 ⁇ A) 680 to improve the adhesion of the Au onto the silicon dioxide layer 660.
  • any photo resist 670 and unwanted Au 680 and Cr 690 are removed by dissolving the photo resist pattern layer 670.
  • the wafer 600 is bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding silicon areas.
  • HMDS hexamethyldisilazane
  • the surface of the biosensor is modified by a surface modification step to prevent non-specific binding.
  • a macromolecule biosensor's surface can be modified by the steps of first cleaning the Au surfaces 680 with concentrated "Piranha" solution (70 vol% H 2 SO 4 , 30 vol% H 2 O 2 ) and thoroughly rinsed with deionized water (dH 2 O).
  • surface modification material such as SAM of biotin-DAD-C12-SH (12-mercapto(8-biotinamide- 3,6-dioxaoctyl)dodecanamide) Roche GmBH, Germany is deposited.
  • a third aspect of the present invention relates to the integration of an entire electrochemical (redox) biosensor onto a single integrated circuit (IC) chip.
  • An integrated circuit (IC) is defined as a tiny wafer of substrate material upon which is etched or imprinted a complex of electronic components and their interconnections.
  • MEMS devices have distinctive properties as a result of small features but the signal level from MEMS-based sensor is relatively low compared to a conventional sensor. Sensitivity can be improved by using an off-chip amplification module but it also increases the noise and the system size.
  • the present inventive entity designed an on- chip amplification circuitry (amplification circuit incorporated on the biosensor chip itself) and detection circuitry (such as providing bias potential, current measurement, sequential control and signal processing) to reduce the inter-chip interference.
  • Both the detection circuitry and the amplification circuitry can be provided by using bipolar transistor (BJT) and/or complementary metal-oxide-semiconductor (CMOS) devices.
  • BJT bipolar transistor
  • an on-chip amplification device is equipped underneath the working electrode, which is typically the largest area of the electrochemical sensor cell.
  • the base region receives the current from the transducer with the current gain beta ⁇ , which ranges from 80-150.
  • beta ⁇ which ranges from 80-150.
  • BJT open-base bipolar
  • the current gain is determined by the length of base region. In vertical BJT, this is a function of ion implementation energy and doping concentration. In horizontal BJT, the length is a function of lithography resolution, ion implementation angle and thermal diffusion. For these reasons the vertical BJT is more reliable in terms of chip-to-chip or in-chip gain uniformity.
  • Fig. 22 shows in cross-section an embodiment of in-chip amplifier circuitry that preferably includes a biosensor comprising three metal electrodes used for electrochemical sensing.
  • the three electrodes are fabricated using integrated circuit (IC) processes and have a relatively large area for interconnection and isolation.
  • the entire biosensor chip 700 can be stacked in two stages (electrochemical sensing stage 730 and bipolar transistor (BJT) amplifying stage 720) and the Au working electrode 710 can act as an electromagnetic shield for the BJT device 720.
  • the working electrode contacts (such as contacts 733, 734, 735, 736, 737 and 738) make contact to a contact interface 740 and also increase the surface area of the working electrode 710 for higher signal.
  • biosensor chip 700 comprises a silicon substrate 744.
  • Substrate 744 includes a base region 743, a collector 742 and an emitter 741.
  • Base region 743 receives electron flux from working electrode 710.
  • Collector 742 is connected to a power source and provides the current gain under certain base conditions for amplification reasons. A resulting current from base 743 and collector 742 is then measured at emitter 741.
  • metal interconnect 750 is connected to signal output (not shown) and emitter 741.
  • Gold working electrode 710 connects to base region 743 through a first layer 760 of silicon dioxide via etching.
  • a second layer of silicon dioxide 770 isolates the BJT 720 and signal line while first layer 760 isolates the three electrodes (working electrode 710, reference electrode 780, and counter electrode 790).
  • the contacts (such as contacts 733, 734, 735, 736, 737 and 738) on working electrode 710 increase the surface area and also form a solid contact with BJT 720.
  • Fig. 24 shows a sectional view of another preferred embodiment of a biosensor unit 800 representing a preferred embodiment of a plurality of biosensors on a substrate
  • the biosensor unit 800 comprises a CMOS and/or BJT layer 830, an electrochemical layer 840 and a reagent containment layer 850, and is generally similar to the Fig. 22 embodiment already described.
  • the third aspect of the present invention is related to a biosensing system that allows for direct integration without chip-to-chip connection of the components in the sensor with other components needed for biological detection.
  • the system does not require an external component (instrument) or requires only a minimum number of external components to constitute a complete system.
  • this all-in-one biosensing system reduces the cost and noise level of biological sensing and simplifies the process of such sensing.
  • Fig. 25 shows an example of how an embodiment of the present biosensor (sensor-chip) can preferably be fabricated by integrated circuit (IC) technology with a CMOS device and/or devices.
  • IC integrated circuit
  • CMOS complementary metal-oxide-semiconductor
  • BJT bipolar junction transistor
  • an integrated circuit (IC) is fabricated on semiconductor substrate 900 with a layer of silicon dioxide 905 with a poly silicon gate 907 on top of a active well region 909.
  • Poly silicon gate 907 is fabricated from an amorphous silicon and it is used for switching the transistor comprises low resistance source 917 and drain 919.
  • an oxide layer (5000A) 920 is next deposited on the substrate 900 for electrical isolation.
  • the silicon dioxide layer 920 is then selectively etched by using lithography and etching methods to form an electric connection hole 940.
  • a conducting plug 950 is then applied into the contact hole 940. Conducting plug 950 is used as interconnection between conductive electrode 960 with low resistance source 917 and/or drain 919.
  • Fig. 29 shows a lift-off process for the fabrication of the necessary electrodes.
  • the electrodes for biosensing are patterned on substrate 900 by using PR5214 photo resist layer 970 on silicon dioxide layer 920.
  • a mask 921 is used to transfer a desired pattern onto photo resist layer 970 by using reverse imaging process (which includes the removal of unwanted photo resist layer 970).
  • a layer 960 of gold Au(200 ⁇ A) and/or Cr(20 ⁇ A) is electron-beam deposited on silicon dioxide layer 920 with the desired photo resist pattern layer 970.
  • any photo resist 970 and unwanted Au/Cr 960 are removed by dissolving the photo resist pattern layer 970. Referring now to Fig.
  • this shows an example of how an embodiment of the present biosensor (sensor-chip) 1000 can preferably be used to detect an ionic molecule, such as iron (Fe).
  • the sensor surface 1010 is not modified and is ready to use after the post fabrication cleaning as shown in the above fabrication embodiment.
  • the analyte used in this embodiment was potassium ferricyanide, K 3 Fe(CN) 6 (329.26 g/mol), which contains an iron atom in the +3 oxidation state (Fe 111 ) in a buffer solution of potassium nitrate, KNO 3 (101.11 g/mol).
  • K 3 Fe(CN) 6 329.26 g/mol
  • the mixed solution 1020 with analytes is applied onto all three electrodes (working 1030, reference 1040 and counter electrode 1050).
  • the volume of the solution 1020 is adjusted so the droplet is confined over all three electrodes by surface tension forces.
  • the Au surfaces were cleaned with acetone, alcohol, and concentrated "Piranha” solution (70 vol% H 2 SO , 30 vol% H 2 O 2 ) and thoroughly rinsed with deionized water (dH 2 O) if the sensor 1000 is to be re-used.
  • Fig. 33 shows a preferred embodiment for macromolecules (e.g., DNA, RNA, protein) detection.
  • the detection of the concentration of macromolecules does include the sensor surface modification of biotin/Streptavidin layer.
  • the sensor surface is modified with a proper biochemical solution to form a streptavidin layer after the post-fabrication cleaning as shown in the surface modification process described above.
  • the amperometric biosensing of pathogen is conducted by first adding 50 ml of lysis reagent (0.4 M NaOH) to a 250 ml sample of bacteria solution and incubated for 5 minute at room temperature.
  • probe solution anchoring and signaling probes
  • 100 ml of probe solution (anchoring and signaling probes) was then added, and the mixture was incubated for 10 min. at 65°C.
  • 5 ⁇ L of the lysed E. coli/probe solution mixture 1120 is then placed on the streptavidin coated working electrode 1130 of the biosensor 1100 and incubated for 10 min. at room temperature.
  • the biosensor 1100 is then washed with biotin wash solution (Kirkegaard and Perry Laboratories, 50-63-06).
  • biotin wash solution Kirkegaard and Perry Laboratories, 50-63-06
  • 5 ⁇ L 1140 of Anti-Fl-POD Anti-fluorescein peroxidase, 150U, Roche Inc., 1 426 346
  • dilutant PBS/0.5%Casein
  • the biosensor is then washed again with wash solution.
  • 10 ⁇ L 1160 of K-blue substrate (Neogen Corp., 300176) is placed on the biosensor in such a way that all three electrodes (working 1130, reference 1140, counter 1150) are covered by the substrate solution 1160.
  • the electrochemical (more specifically the redox) measurements are immediately taken.
  • Amperometric current vs. time is measured using a CH Instruments 660A Electrochemical Workstation with a picoamp booster and faraday cage. Samples on the biosensor are to be measured sequentially. The voltage should be fixed at -0.1V (vs. reference), and a cathodic current (amperometric signal) reading should be taken in 20 seconds because at 20 seconds, the current values should reach steady-state.
  • Cell concentration (cell number) is determined by using serial dilutions and culture plate counting.
  • the Au surfaces should then be cleaned with acetone, alcohol, and concentrated "Piranha” solution (70 vol% H 2 SO 4 , 30 vol% H 2 O 2 ) and thoroughly rinsed with deionized water (dH 2 O) if the sensor 1100 is to be re-used.
  • the biosensor is calibrated before actual sample detection.
  • the biosensor is preferably calibrated with a calibrating solution that contain a known amount of target analyte(s) and another calibrating solution that contains an undetectable amount (e.g., none) of the target analyte(s).
  • the biosensor may be calibrated with a plurality of calibrating solutions. Each of the plurality of calibrating solutions respectively contains a known amount of the target analyte.
  • the calibrating solutions contain the target analytes at different level of concentrations.
  • the reference signal and/or signals are then compared with the measured signal from a sample solution to determine the presence and quantity of the analyte in the sample reagent.
  • the determination of the presence and quantity (e.g., concentration or absolute amount) of the analyte(s) can be determined by conventional biological interpolation methods. Appendix A provides additional material to further characterize the present invention.
  • Ruzgas T., Gorton, L., Emne'us, J., Marko-Varga, G., 1995. Kinetic models of horseradish peroxidase action on a graphite electrode. J. Electroanal. Chem. 391, 41-49.
  • FIG.1 A reusable DNA sensor array for rapid biological agent detection has been fabricated on a silicon chip.
  • the DNA-based probes target the DNA/RNA sequence of the analyte instead of indirect probing using antibodies.
  • the sensitivity is greatly enhanced by combining the hybridization event with a signal enzyme.
  • the formation of the self-assembled monolayer sensor surface, in-situ DNA hybridization, signal measuring and the sensor regeneration can be performed within 40 minutes. Even without using the PCR, as low as 1000 Escherichia coli (E. coli) cells through 16s rRNA can be detected using this sensor array.
  • FIG.2 The dimension of each sensor area ranges from 25 mm 2 to 160 ⁇ m 2 .
  • Electrochemical sensor Microelectromechanical system (MEMS), Self-assembled monolayers (SAMs), DNA hybridization
  • the enzyme-based electrochemical biosensor is primarily motivated by the need for a highly sensitive and selective protocol capable of rapid monitoring the concentration of bacteria, virus or various biological species for field use. Such a protocol would operate remotely, and would be fully automated, compact, and robust [1]. Therefore the electrochemical transducer with minimum power consumption and smaller size is preferred over the optical system [2].
  • High specificity can be achieved by using DNA hybridization to reduce false positive and false negative signals.
  • DNA electrochemical biosensors have been previously reported [3,4] using graphite or carbon electrodes. Carbon-based electrodes, however, are generally not adaptable to MEMS technology when small ( ⁇ m) dimensions are needed.
  • Au is used as electrodes, with a protein self-assembled monolayer (SAM) to capture the E. coli rRNA.
  • SAM protein self-assembled monolayer
  • An enzyme is used as a biological amplifier in this study to gain the high sensitivity without PCR.
  • a sensitivity and specificity check for E. coli MC4100 versus Bordetella SB54
  • HRP is one of the most widely used enzymes for analytical purposes and biosensors application [5]. Enzymatic amplification occurs due to a high turnover number in reactions that can be detected electrochemically.
  • HRP horseradish peroxidase
  • TMB 3,3'5,5'-tetramethylbenzidine
  • Fig.3 The electron transfer at the electrode surface is measured amperometrically to represent the number of the enzymes immobilized by DNA hybridization though the 16s ribosomal RNA of the target cell. Therefore, the output current is proportional to the number of the target cells in the solution.
  • a three-electrode electrochemical cell is constructed with a micro-fabricated reaction well for the working electrode (Fig.4a).
  • Gold Au
  • Fig.4b Gold
  • the surrounding Si surface is modified to be hydrophobic.
  • the three dimensional reaction well along with the hydrophobic nature of the surrounding area allows a liquid droplet to be well contained in the working electrode. This design minimizes non-specific binding of biomolecules to other areas of the sensor chip.
  • the Au working electrode has a monolayer of streptavidin immobilized on the surface via a thiolated-biotin SAM or through direct protein adsorption onto the gold.
  • Fig.4c A sample solution containing E.
  • coli is treated with lysis buffer, the target DNA/RNA from the E. coli cells are hybridized with both an anchoring ssDNA probe and a labelling ssDNA probe at annealing temperature ( ⁇ 65°C) in the presence of cell debris.
  • the anchoring probe (conjugated to biotin) and the labelling probe (conjugated to fluorescein) recognize two distinct conservative sequences, therefore, the hybrid forms only with the specific gene segment from the target bio-agent.
  • the oligonucleic hybrid is then immobilized through biotin-streptavidin binding onto the working electrode and unbound components are washed away.
  • a HRP-linked anti-fluorescein antibody is then loaded onto each hybrid (Fig.4e). After addition of substrate, enzymatic reaction causes a current signal which is measured amperometrically using the three-electrode cell.
  • Fig.4f The entire protocol is completed in 40 minutes.
  • SPR streptavidin SAM
  • AFM atomic force microscopy
  • Atomic Force Microscopy was performed in the contact mode using ultralever tips with a force of 5.0nN.
  • the Au substrate was prepared following the method of Wagner, et al. [7].
  • the bare Au has topographic features ⁇ l ⁇ A (Fig. ⁇ a) and contact AFM of a "full" monolayer showed a smooth surface with evidence of protein dragging (Fig. ⁇ b).
  • the height of protein islands on a "partial" monolayer was -45 A, consistent with the dimensions of the streptavidin protein.
  • the AFM observations confirm the SPR results that only one monolayer was deposited.
  • Fig. of SA ormation on u. a are u on y u coverage w t prote n dragging (c) partial coverage with protein islands.
  • This MEMS based DNA sensor utilizes a standard three-electrode electrochemical cell configuration with novel micro fabricated structure design to minimize non-specific binding.
  • the sensor module is easily to be adapted to various protocols and can be used for rapid detection of macromolecules (DNA, RNA) from targets such as uropathogenic Escherichia coli (E. coli) in urine and microorganisms causing otitis media (middle ear infection).
  • E. coli Escherichia coli
  • otitis media middle ear infection
  • Less than 10 5 E. coli cells can be detected from the urine sample of a patient with urine tract infection.
  • the sensitivity is enhanced by appropriate sensor characterization and surface modification.
  • the total detection time including sample preparation can be reduced to 25 minutes by using a POD conjugated oligonucleotide.
  • An electrochemical cell must consist of at least two electrodes (working electrode, reference electrode) and an electrolyte.
  • An electrochemical electrode is an interface at which the mechanism of charge transfer changes between electronic transport and ionic transport.
  • An electrolyte is a medium through which charge transfer can take place by the movement of ions.
  • Electrochemical detection requires a second unvarying potential supplied by a reference electrode, forming a half battery.
  • a typical reference electrode is Ag/AgCl.
  • Most MEMS based electrochemical sensors focus on micro fabrication of only the working electrode. This work investigates the characteristics and application of a MEMS based electrochemical sensor on silicon fabricated by standard MEMS processes. Testing of clinical urine samples with DNA hybridization on the electrochemical sensor demonstrates that MEMS based sensor on silicon can be used for biomedical detection.
  • RNA/DNA Nucleic acid molecules from chemically disrupted target cells are hybridized with both an anchoring ssDNA probe (conjugated to biotin) and a labeling ssDNA probe (conjugated to fluorescein) in the presence of cell debris.
  • anchoring ssDNA probe conjuggated to biotin
  • labeling ssDNA probe conjuggated to fluorescein
  • Fig.2a These two probes recognize two distinct conservative sequences, and therefore, the hybrid forms only with the specific genetic segment from the target bio-agent.
  • the oligonucleic hybrid is then immobilized through biotin-streptavidin binding onto the working electrode and unbound components are washed away.
  • a peroxidase (POD)-linked anti- fluorescein antibody is then loaded onto each hybrid.
  • enzymatic reaction causes a current signal which is measured amperometrically from the REDOX reaction shown in Fig.3.
  • the entire protocol can be completed in 40 minutes and the sensor reactivated after a cleaning process.
  • Cyclic voltammetry for reversible one-electron transfer of ferrocene is characterized by a peak separation of ⁇ 57 mV between the anodic and cathodic peaks, the same peak maximum, and a linear relationship between peak current vs. [scan rate] [4].
  • voltammograms of ferrocene using MEMS based electrochemical sensor at different scan rates show the expected behavior and a plot of the peak current at
  • the sensor surface has a protein self-assembled monolayer to capture the target RNA/DNA onto the working electrode, and the SAMs have been characterized by atomic force microscopy (AFM) and surface plasmon resonance (SPR).
  • AFM atomic force microscopy
  • SPR surface plasmon resonance
  • the signal-to-noise can be improved significantly by appropriate surface modifications to reduce non-specific binding.
  • the first approach is to introduce a surface blocking protein to eliminate potential bonding sites for non-specific binding.
  • Fig 6 The second approach is adding a hydrophobic modification of the periphery region with silanation treatment or thin-film coatings of Teflon® or polyimide.
  • Protocol simplification Sample preparation is the most time consuming step for most biosensors. Any additional protocol steps will require more fluidic devices and a longer detection time.
  • a simplified protocol is shown in Fig.9 whereby the step of the enzyme loading with POD conjugation onto detector DNA probe is eliminated. The hybridization condition was controlled to retain the enzymatic activity of POD after heating to 65°C. The protocol can then be shortened fro m 40 to 25 minutes while reducing the required number of reagents by one.
  • Micro DNA sensor can be used for electrochemical detection without utilizing a conventional reference electrode because the bias potential is reasonable low (-0.1V) and the detection is short enough (20 seconds) to avoid the accumulation of charge at auxiliary electrode. This was confirmed with voltammograms of ferrocene and POD solution.
  • micro DNA sensor can be used for electrochemical detection of pathogens such as E. coli.
  • pathogens such as E. coli.
  • the Au/Au/Au three electrode cells patterned on silicon by MEMS technology were successfully used in amperometric measurement of enzymatic reactions. These sensors show promise that they can be inco ⁇ orated into micro total analysis system ( ⁇ -TAS) or "Lab on a Chip”.
  • a micro DNA sensor was fabricated that can detect enzymatic reaction amperometrically and was characterized by using cyclic voltammetry with ferrocene and POD to demonstrate the capability of conducting electrochemical detection without a conventional Ag/AgCl electrode. Noise reduction was accomplished by surface modification and introducing blocking protein.
  • the structural design of a well in the working electrode and surface treatment on silicon substrate enable reagent confinement over the working electrode. The reagent-electrode contact can be controlled to reduce non- specific binding, not possible with a conventional beaker setup. Urine sample testing shows this DNA sensor is suitable for clinical diagnosis with a short detection time and smaller system size.
  • E. coli Escherichia coli
  • MEMS microelectromechanical systems
  • SAMS self-assembled monolayers
  • DNA hybridization DNA hybridization
  • enzyme amplification enzyme amplification
  • MEMS Microelectromechanical systems
  • SAMs self-assembled monolayers
  • amperometric detection Escherichia coli bacteria
  • DNA hybridization
  • MEMS Microelectromechanical systems
  • MEMS technology provides transducers to perform sensing and actuation in various engineering applications.
  • the significance of MEMS technology is that it makes possible mechanical parts of micron size that can be integrated with electronics and batch fabricated in large quantities.
  • MEMS devices are fabricated through the process of micromachining, a batch production process employing lithography. Micromachining relies heavily on the use of lithographic methods to create 3 -dimensional structures using pre-designed resist patterns (or masks) and then selectively etching the undesirable parts away (Ho and Tai, 1996, 1998).
  • MEMS is an enabling technology for making miniaturized devices, and in this work, we integrate MEMS technology with biosensing methods to detect E. coli bacteria.
  • One of the most effective means of achieving high specificity is to detect the bacteria's genetic material (e.g. rRNA, mRNA, denatured DNA).
  • a single- stranded DNA (ssDNA) probe whose sequence is complementary only to the target bacteria's rRNA or ssDNA
  • monitoring the hybridization event allows selective sensing of target cells.
  • ssDNA single- stranded DNA
  • coupling the hybridization event with an enzymatic reaction leads to signal amplification, as each substrate-to-product turnover contributes to the overall signal.
  • Bioassays to detect DNA hybridization that are amplified by enzymatic reaction can still be completed within a reasonably short time.
  • the miniaturization and portability inherent in electrochemical probes make them excellent candidates for incorporation in MEMS devices.
  • E. coli Escherichia coli
  • coli using screen-printed carbon electrodes on disposable test strips are also available (Andcare Inc., Durham, NC). Screen-printing has the advantage of low cost, but achieving high dimensional precision is not easy .
  • lithography thin films of a wide range of materials, including metals (Au, Ag) and carbon, can be accurately patterned in ⁇ m size dimensions.
  • SAMs is an elegant method of selectively immobilizing molecules on MEMS surfaces.
  • a layer of silicon dioxide (SiO 2 , lOOOA) was deposited on a bare Si wafer (prime grade, p-type ⁇ 100>, thickness 500-550 ⁇ m) and served as a pad layer underneath the silicon nitride (Si 3 N , 1000 A) to release stress and improve adhesion.
  • MEMS arrays were fabricated with working electrodes of 3.6mm x 3.6 mm etched to form wells of 350 ⁇ m depth.
  • the nitride wafer was patterned and bulk etched using KOH along [111] and [100] crystal planes, and depth of the well was controlled by KOH etching time and temperature.
  • the lOO ⁇ m wide auxiliary and reference electrodes are separated from their corresponding working electrode by 200 ⁇ m.
  • Figure 1 shows a schematic of the pattern used in generating the MEMS array.
  • the nitride and oxide were removed by HF etching to release internal stress, and another oxide layer (5000A) was deposited for electrical isolation.
  • Electrodes were patterned by PR5214 photo resist reverse imaging and lift-off process with e-beam deposition of Au(200 ⁇ A)/Cr(20 ⁇ A).
  • the wafer was bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding Si areas.
  • HMDS hexamethyldisilazane
  • streptavidin monolayers on Au Three different methods were used to deposit streptavidin monolayers on Au: 1) directly adsorbing streptavidin on bare Au, 2) depositing a SAM of a biotinylated thiol, biotin-DAD-C12-SH, and subsequently binding streptavidin, 3) depositing a SAM of a biotinylated disulfide, biotin-HPDP, and subsequently binding streptavidin.
  • the Au surfaces were cleaned with concentrated "Piranha" solution (70 vol% H 2 SO , 30 vol% H 2 O 2 ) and thoroughly rinsed with deionized water.
  • streptavidin For depositing streptavidin on bare Au, a solution of 1.0 mg/ml streptavidin (Sigma Chemical Co., S0677) in 0.02M Na phosphate buffer, 0.15M NaCl, pH 7.2, was placed on the surface, allowed to stand for 10 minutes, and rinsed with deionized water.
  • streptavidin Sigma Chemical Co., S0677
  • 0.02M Na phosphate buffer 0.15M NaCl, pH 7.2
  • the assay protocol was conducted as follows: 1) 50 ⁇ l of lysis reagent (Andcare Inc., 4002-11) was added to a 250 ⁇ l sample of bacteria in culture media and incubated for 5 min. at room temperature, 2) 100 ⁇ l of probe solution (Andcare Inc., 4002-13) was then added and the mixture was incubated for 10 min. at 65°C, 3) 5 ⁇ l of the lysed E. coli/probe solution mixture was placed on the streptavidin coated working electrode of the MEMS detector array and incubated for 10 min.
  • the MEMS detector array was washed with biotin wash solution (Kirkegaard and Perry Laboratories, 50-63-06), 5) 5 ⁇ l of Anti-Fl-POD (Anti-fluorescein peroxidase, 150U, Roche Inc., 1 426 346), diluted to 0.75U/ml or 0.15 U/ml with diluant (Andcare Inc., 4002-14) was placed on the working electrode and incubated for 10 minutes at room temperature, 6) the MEMS array chip was washed again with wash solution, 7) 10 ⁇ l of K-blue substrate (Neogen Corp., 300176) was placed on the detector array in such a way that all three electrodes (working, auxiliary, reference) were covered by the substrate solution, and 8) electrochemical measurements were immediately taken.
  • biotin wash solution Karlegaard and Perry Laboratories, 50-63-06
  • Anti-Fl-POD Anti-fluorescein peroxidase, 150U, Roche Inc., 1 426 346), diluted
  • the performance of the detector depends heavily on the properties of the immobilized streptavidin monolayer.
  • SPR surface plasmon resonance
  • AFM atomic force microscopy
  • biotin SAM was deposited on the bare Au chips as previously described before the SPR experiments. For best results, new chips were cleaned with diluted H SO /H 2 O 2 solution for -2 min.
  • MEMS Microelectromechanical system
  • Figure 2 shows a photograph of the MEMS detector array. Sixteen working electrodes with their corresponding auxiliary and reference electrodes were patterned in a 2.8 cm x 2.8 cm area. The detector array was fully reusable as the surface can be cleaned using H 2 SO /H 2 O 2 solutions. We have reused the same MEMS detector array multiple times by appropriately cleaning the surface and redepositing the SAMs on the working electrode. 3.2 E. coli Detection Using DNA Hybridization
  • E. coli detection is based on DNA hybridization followed by enzymatic reaction.
  • a schematic illustrating the electrode surface is shown in Figure 3.
  • a streptavidin monolayer is immobilized on the Au working electrode surface to capture the rRNA from E. coli.
  • Two ssDNA segments are used in this system.
  • the capture ssDNA which is conjugated to biotin for streptavidin binding, hybridizes to one end of the E. coli rRNA.
  • the detector ssDNA which is conjugated to fluorescein for binding to anti- fluorescein linked to the enzyme peroxidase (POD), hybridizes to the other end of the E. coli rRNA.
  • POD peroxidase
  • the capture and detector ssDNA recognize two distinct conservative sequences, and therefore, the hybrid forms only with the specific gene segment from E. coli.
  • the oligonucleic hybrid is immobilized through biotin-streptavidin binding onto the Au working electrode and unbound components are washed away. Streptavidin binds biotin with unusually high affinity (K d -10 "15 M) (Weber, et. al., 1989).
  • substrate After loading the POD onto the hybrid (through Anti-Fl-fluorescein binding), substrate is added and enzymatic reaction is detected amperometrically.
  • the substrate solution contains both the substrate H 2 O 2 and a mediator, 3,3',5,5'tetramethylbenzidine (TMB).
  • TMB 3,3',5,5'tetramethylbenzidine
  • a streptavidin monolayer was deposited on bare Au via protein adsorption.
  • a SAM of biotin was deposited on the Au using a biotinylated thiol and streptavidin was subsequently bound to the biotin.
  • a SAM of biotin was deposited on the Au using a biotinylated disulfide and streptavidin was subsequently bound to the biotin.
  • the streptavidin monolayers were characterized using both surface plasmon resonance (SPR) and atomic force microscopy (AFM).
  • SPR has been demonstrated to be a viable technique for monitoring interactions of molecules with metallic (Au, Ag) thin films at the solution-metal interface. This technique can be used to estimate the thickness of a deposited layer as well as to measure the kinetics of association and dissociation (Haussling, et al., 1991, Spinke, et al., 1993, Sigal, et al., 1996, Rao, et al., 1999, Jung, et al., 1999).
  • streptavidin has dimensions of - 55 x 45 x 50 A (Darst, et al., 1991).
  • a full monolayer of streptavidin has an expected density of -2.8 ng/mm 2 , calculated based on a 2-dimensional crystalline monolayer (Jung, et al., 1999, Darst, et al., 1991).
  • streptavidin can be desorbed, i.e. whether streptavidin can be dissociated from the bare Au or from binding to biotin to regenerate the surface.
  • Table I lists the loss in SPR signal (RU) after treatment with various reagents that are known to dissociate protein-ligand binding and/or denature proteins. As seen in Table I, only 8M urea, 0.5% SDS, and 0.1M NaOH were somewhat effective in desorbing streptavidin from the surface. 1.0M KC1, 0.1M HC1, and 40% formamide were not effective. Streptavidin could not be completely desorbed by any of the reagents, and some protein remained after subjecting the surface to all reagents. These results show that streptavidin had rather good binding to both the biotin SAM as well as to bare Au and that the streptavidin monolayers were relatively stable.
  • streptavidin immobilized via the biotinylated thiol to Au was the best condition for E. coli detection.
  • biotin-thiol SAM we obtained good signals for the E. coli while achieving a low baseline signal from the Bordetella.
  • current signals for the E. coli were significantly lower, while the baseline was the same as that for the biotin-thiol/streptavidin.
  • streptavidin directly adsorbed to Au the signal from the Bordetella was much higher, indicating a higher level of non-specific binding of POD to the surface.
  • MEMS technology enables an array of multiple three-electrode "cells" to be deposited on a Si wafer, and the MEMS detector array we have described is fully reusable as the SAMs can be removed and the Au surfaces regenerated with appropriate cleaning.
  • SAMs provide an effective means of functionalizing the Au working electrode to immobilize capture biomolecules, for example, streptavidin.
  • DNA hybridization permits high specificity for pathogenic bacteria as the sequence of the ssDNA probes can be carefully selected to complement only the target. Coupling the hybridization event with an enzymatic reaction provides signal amplification and enhances the sensitivity. Finally, by using electrochemical transduction, a miniaturized portable system with minimum power consumption can be developed.
  • Rapid detection and a portable instrument is desirable for pathogen sensing.
  • detection can be achieved within -40 minutes using this system. Due to the small dimensions and small sample volumes, it is possible to further reduce the assay time by reducing the incubation times (10 minutes) currently used for DNA hybridization and enzyme binding.
  • a distinct advantage of MEMS is the ability to use very small volumes (a few ⁇ l) and electrode surface areas (currently 0.13 cm 2 for the working electrode, ⁇ 0.02 cm for the auxiliary and reference electrodes). Our results show that as few as -10 3 cells can be detected using this system without polymerase chain reaction (PCR).
  • Streptavidin immobilized via direct adso ⁇ tion to Au resulted in significantly higher nonspecific binding of the POD enzyme to the working electrode.
  • Protein adso ⁇ tion to Au has been well known as colloidal Au particles attached to various proteins (e.g. streptavidin, immuno globulins) are commercially available (Nanoprobes, Inc., Yaphank, NY). Streptavidin does not contain cysteine or methionine residues (Weber, et. al., 1989) and therefore, does not attach to Au via an Au-S bond. Protein adso ⁇ tion to Au can occur via interaction of carboxylate groups with Au (Ooka, et. al., 1999) and is the likely mechanism for streptavidin-Au attachment.
  • streptavidin monolayer can be attached to Au via direct adso ⁇ tion, whether self-assembly or molecular ordering occurs is questionable.
  • SPR deso ⁇ tion experiments showed the streptavidin-Au attachment to be as robust as streptavidin-biotin binding, i.e. the amount of streptavidin removed due to urea, SDS, and NaOH were similar for streptavidin directly adsorbed on Au as compared to streptavidin attached to biotin.
  • sample solutions contained oligonucleotides as well as cell debris from the lysed E. coli.
  • Our data suggests that the presence of other proteins and biomolecules accelerated the deso ⁇ tion of streptavidin from Au, leading to increased non-specific binding of the enzyme POD to the surface.
  • E. coli bacteria were successfully detected by inco ⁇ orating MEMS with SAMs, DNA hybridization, and enzyme amplification.
  • MEMS-based detection system that is specific for E. coli and capable of detecting 1000 cells without PCR.
  • the process time can be 40 minutes or less.
  • the assay can be conducted with solution volumes on the order of a few microliters.

Abstract

A microelectromechanical system (MEMS) and integrated circuit based biosensor (210) capable of sensing or detecting various ionic molecules and macromolecules (DNA, RNA or protein). The MEMS based biosensor (210) may utilize a hybridization and enzyme amplification scheme and an electrochemical detection scheme for sensitivity improvement and system miniaturization. The biosensor or biosensors (210) are incorporated on a single substrate (200). Preferably, the biosensor system comprises at least two electrodes. The electrodes may comprise a working electrode (220), a reference electrode (240), and a counter (auxiliary) electrode (230). The biosensor or biosensors (210) also provide an apparatus and method for confinement of reagent and/or solution in the biosensor or biosensors (210) using surface tension at small scale. The confinement system provides controlled contacts between the reagent(s) and/or solution(s) with the components (i.e., electrodes) of the biosensor or biosensors (210) using controllable surface properties and surface tension forces. The confinement system allows for incorporation of the biosensor or biosensors (210) into a portable or handheld device and is immune to shaking and/or flipping. The invention also provides for a biosensor (210) and/or sensors that are integrated with integrated circuit (IC) technologies. Preferably, the entire sensor system or systems are fabricated on a single IC substrate (200) or chip and no external component and/or instrument is required for a complete detection system or systems. Preferably, the sensor system or systems are fabricated using the IC process on a silicon substrate (200).

Description

DESCRIPTION Biological Identification System With Integrated Sensor Chip
This application claims the benefit of U.S. Provisional Patent Application Serial No. 60/201,603, filed May 3, 2000.
Statement Regarding Federally Sponsored Research Or Development
This invention was made with Government support under Grant No. N66001-96- C-8632 awarded by the Department of the Navy. The Government has certain rights in this invention.
Field Of The Invention The field of the present invention relates generally to apparatus and methods for sensing or detecting various target analytes, including ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein) and in particular, to biosensors, methods of using the biosensors, and methods for making the biosensors.
Background Of The Invention
A variety of biosensors have been developed for the detection of biological material, such as pathogenic bacteria. Conventional methods for detecting bacteria usually involve a morphological evaluation of the organisms and rely on (or often require) growing the number of organisms needed for such an evaluation; such methods are time consuming and are typically impractical under field conditions. The need for rapid detection as well as portability has led to the development of systems that couple pathogen recognition with signal transduction. Both optical and electrochemical detection of bacteria have been reported (Ivnitski, et. al., 1999, T. Wang, et. al., 2000). (Full citations to the literature cited herein are given at the end of the specification.) However, the present inventive entity has determined that electrochemical methods have an advantage in that they are more amenable to miniaturization.
Requirements for an ideal detector include high specificity and high sensitivity using a protocol that can be completed in a relatively short time. Moreover, systems that can be miniaturized and automated offer a significant advantage over current technology, especially if detection is needed in the field.
The electrochemical methods use the principle of electrical circuit completion. To complete the electrical circuit, a counter electrode is used to provide a return path to the sample solution or reagent and a reference electrode is used as a reference point against which the potential of another electrode or electrodes are determined (typically that of the working electrode or measuring electrode). Since this contact must be provided by electrochemical means, i.e. a metal electrode immersed in a chemical solution or reagent, it is impossible to avoid generating an electrical potential in series with the potential developed by the electrode. The conventional theory in the electrochemical methods is that it is essential for the reference electrode potential to be very stable and not be affected by chemical changes in the solution. Thus, silver/silver chloride reference electrodes, which provide a very stable reference potential, are the most common type of electrode used for reference electrodes today. Referring to Fig. 1, the typical silver/silver chloride reference electrode 10 contains a chloridised silver wire 1 (a layer of silver chloride coated on silver wire) immersed in a solution 5 of potassium chloride (3.5M KC1) saturated with silver chloride (AgCl). This internal filling solution 5 slowly seeps out of the electrode 10 through a porous ceramic junction 20 and acts as an electrical connection between the reference element 1 and the sample. Potassium chloride is used because it is inexpensive and does not normally interfere with the measurement. The solution 5 also includes silver chloride to prevent dissolution of the coating on the reference element 1. It is therefore necessary to maintain the level of solution 5 in the electrode 10 using a filling solution hole 40.
The present inventive entity, however, has determined that robust and precise biological detection can be achieved using electrochemical methods, and more precisely using redox (reduction-oxidation) methods, without the need of a two-layered reference electrode (e.g., silver chloride coated on silver) having a known reference electrode potential described above.
Referring now to Fig. 2, the electrochemical methods also required a potentiostat 50, which is a control amplifier with the test cell placed in the feedback loop. The objective is to control the potential difference between a test electrode (working electrode) 60 and a reference electrode 70 by the application of a current via the third, auxiliary electrode (counter electrode) 80. In practice a fairly good potentiostat may be built using a minimum of components. In the circuit shown here, 90A and 90B are 1.22V bandgap reference diodes connected between the positive and negative power rails. Potentiometer 100 is then used to set the required cell polarization, applied to the non-inverting input of the main amplifier 110. The working electrode 60 connects to the ground of the circuit and the reference electrode 70 to the inverting input of the main amplifier 110. In order to boost the output capability somewhat (most operational amplifiers are limited to about 20 mA and do not tolerate capacitative loads well) a unity gain buffer amplifier 120 is used. Preferably, gain buffer amplifier 120 has a much higher bandwidth than amplifier 110, otherwise the circuit is likely to oscillate when driving a capacitative cell. The output of the buffer amplifier 120 connects to the auxiliary electrode 80 via a current measuring resistor 140. Differential amplifier 130 is then used to measure the voltage drop across this resistor 140 and to convert it to a ground referenced output voltage.
Microelectromechanical systems (MEMS) technology provides transducers to perform sensing and actuation in various engineering applications. The significance of MEMS technology is that it makes possible mechanical parts of micron size that can be integrated with electronics and batch fabricated in large quantities. MEMS devices are fabricated through the process of micromachining, a batch production process employing lithography. Micromachining relies heavily on the use of lithographic methods to create 3-dimensional structures using pre-designed resist patterns (masks) (Ho and Tai, 1996, 1998). MEMS is one suitable technology for making micro fabricated devices or aspects thereof. Microfabricated devices are generally defined as devices fabricated by using MEMS and/or integrated circuit (IC) technology. An integrated circuit (IC) is defined as a tiny chip of substrate material upon which is etched or imprinted a complex of electronic components and their interconnections.
The present inventive entity has determined that miniaturization and portability features are inherent in electrochemical methods make these methods excellent candidates for integration into MEMS devices, and, further that it would be advantageous to integrate MEMS technology with biosensing methods to detect various ionic molecules and macromolecules (DNA, RNA or protein).
The present inventive entity has also determined that in the area of biosensing of bacteria, one of the most effective means of achieving high specificity is to detect the bacteria's genetic material (e.g. rRNA, mRNA, denatured DNA). By choosing a single- stranded DNA (ssDNA) probe whose sequence is complementary only to the target bacteria's rRNA or ssDNA, monitoring the hybridization event allows selective sensing of target cells. To maximize sensitivity, coupling the hybridization event with an enzymatic reaction leads to signal amplification, as each substrate-to-product turnover contributes to the overall signal. Biosensing to detect DNA hybridizations that are amplified by enzymatic reaction can still be completed within a reasonably short time according to the invention.
Thus, a prototype amperometric detector for Escherichia coli (E. coli) has been developed based on the above determinations and technologies (Chen, et. al., 2000, Gau, et. al., 2000). The technologies of MEMS, self-assembled monolayers (SAMs), DNA hybridization, and enzyme amplification all contribute to the design of a miniaturized, specific, and sensitive E. coli detector. DNA electrochemical probes have been reported previously (Wang, et al., 1997, Marrazza, et al., 1999), with graphite or carbon electrodes typically used. Commercial units for amperometric detection of DNA from E. coli using screen-printed carbon electrodes on disposable test strips are also available. Screen- printing has the advantage of low cost; however, achieving high dimensional precision is not easy. Thus, the present inventive entity has determined a need to develop a method of using lithography to accurately pattern in μm size dimensions a wide range of materials such as metals (e.g. Au, Ag) and carbon. Moreover, utilizing a surface modification, such as self-assembly monolayer (SAM) of biotin-DAD-C12-SH dodecanamide, is a preferred method of selectively immobilizing molecules on MEMS surfaces. The formation of SAMs on Au, Ag and other metals has been well studied (Revell, et al., 1998, Motesharei, et al., 1998, Xia, et al., 1998, Lahiri, et al., 1999), and proteins and other biomolecules can be easily immobilized onto surfaces such as Au using SAMs (Ostuni, et al., 1999, Kane, et al., 1999, Spinke, et al., 1993, Haussling, et al., 1991). Amperometric methods using SAMs on electrodes have demonstrated the ability to detect target analytes successfully (Sun, et al., 1998, Hou, et al., 1998, Murthy, et al., 1998).
In the instant E. coli detection system, the present invention identifies and takes advantage of certain benefits inherent in each technology. Using DNA hybridization and enzyme amplification, the present invention achieves the required specificity and sensitivity. Using MEMS and SAMs, the present inventive entity fabricated a miniaturized system that can be developed into a portable instrument. Finally, the invention demonstrates that the present detection system is applicable to a broad range of pathogenic bacteria. For example, the detection module and assay protocol can be adapted to detect uropathogenic E. coli and identify microorganisms causing otitis media (middle ear infection).
Accordingly, it is an object of the invention to provide novel biosensors, novel methods of using the biosensors, and novel methods for making the biosensors. This object is solved by the combination of the features of the main claim, and the sub-claims disclose further advantageous embodiments of the invention.
Summary of the Invention
This summary of the invention does not necessarily describe all necessary features detailed in the specification, and the invention may also reside in a sub-combination of features described here and elsewhere in the specification.
A first aspect of the present invention provides an apparatus and method for sensing or detecting various target analytes, including especially macromolecules (e.g.
DNA, RNA or protein) and ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) using a biosensor incorporated on a single substrate (silicon, glass, plastic, etc.). The substrated biosensor system comprises at least two electrodes that are typically fabricated together as a single series microfabrication process step on the substrate. However, successive microfabrication steps may also be employed to fabricate the system. In a preferred embodiment, the electrodes are all made out of pure metal (as opposed to the Ag/AgCl electrodes of the prior art, for exmple). In another preferred embodiment, the substrated biosensor system comprises a working electrode, a reference electrode and a counter (auxiliary) electrode. Preferably the substrated biosensor system has the same electrochemical performance as a conventional electrochemical biosensor. In addition, the substrated biosensor system should be compatible with integrated circuit (IC) and/or MEMS fabrication processes and be capable of being constructed in a small area.
A second aspect of the present invention provides an apparatus and method for confinement of reagent and/or solution in a biosensor using surface tension at small scale. The reagent and/or solution contain the necessary electrolytes and/or analytes needed for biological sensing. In a preferred embodiment, the apparatus and method in the present invention allow for each electrode used for sensing or detecting various ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein) to be in selective contact with the reagent and/or solution when the electrolytes and/or analytes are needed by using controllable surface properties and surface tension forces at small scale.
In another preferred embodiment, the apparatus and method for confinement of reagent and/or solution (and thus the electrolytes and/or analytes in the reagent and/or solution) using surface tension is incorporated with a portable or handheld device and is immune to shaking of the device. In addition, the reagent and/or solution should be held firmly in position by the biosensor using surface tension even when the biosensor is flipped upside down.
A third aspect of the present invention provides an apparatus and method for integrating the components of an electrochemical sensor and/or sensors (e.g. electrodes) and additional required electronic circuit components (e.g. amplifiers) in an electrochemical sensor or sensors with integrated circuit (IC) technologies. The entire sensor system and/or systems can be incorporated on a single IC substrate or chip, such as a single semiconductor (e.g. silicon or gallium arsenide) substrate or chip. Preferably, no or much fewer external components and/or instruments are required to complete the system or systems. The sensor and/or sensors are preferably fabricated using the IC process.
In any of the embodiments used for macromolecule electrochemical detection, a preferred feature of the invention is to modify the surface on at least one of the electrodes. Preferably, the surface is modified for anchoring macromolecules on the surface. Preferably, the surface is modified using a self-assembly monolayer (SAM) such as biotin- streptavidin. Preferably, the SAM is placed on the surface of a working electrode in an electrochemical sensor. Optionally, materials such as sol gel and/or carbon paste may be used to modify the surface (as a replacement for SAM or in combination with SAM).
The above "summary of the invention" presents only examples and not limitations. Further embodiments as well as sub-combinations, modifications, variations and enhancements of the invention will become apparent in the detailed description of the invention, which follows herein.
Brief Description of the Drawings
In the drawings, wherein the same reference number indicates the same element throughout the several views:
Fig. 1 is a schematic of an embodiment of a conventional reference electrode. Fig. 2 is a schematic of an embodiment of a potentiostat used in the electrochemical methods.
Fig. 3 is a schematic of an embodiment of a plurality of electrochemical biosensors on a circular substrate wafer according to the present invention.
Fig. 4 is a schematic of alternative embodiment of a plurality of electrochemical biosensors on a square substrate according to the present invention. Fig. 5 is a schematic of another embodiment of an electrochemical biosensor on a substrate according to the present invention.
Fig. 6 is a graph of Cyclic Voltammetry (CV) scan potential over time. Fig. 7 is a one-cycle Cyclic Voltammetry (current vs. bias potential) taken by a biosensor according to the present invention. Fig. 8. is a Cyclic Voltammetry (current vs. bias potential) at different scan rate taken by a biosensor according to the present invention.
Fig. 9. is a graph of the square root of scan rate vs. peak current taken by a biosensor according to the present invention.
Fig. 10 is a Cyclic Voltammetry (current vs. bias potential) on a plurality of cycles at constant scan rate taken by a biosensor according to the present invention.
Figs 11(a), (b) and (c) are diagrams showing reagent and/or solution confinement by surface tension and treatment according to the present invention.
Fig. 12 is a diagram showing reagent and/or solution confinement selectively over only a working electrode on a biosensor according to the present invention. Fig. 13 is a diagram showing reagent and/or solution confinement over all electrodes on a biosensor according to the present invention. Fig. 14 is a side view diagram showing the first step of how an embodiment of a biosensor of the present invention can be fabricated.
Fig. 15 is a diagram showing the second step of how the biosensor can be fabricated. Fig. 16 is a diagram showing the third step of how the biosensor can be fabricated.
Fig. 17 is a diagram showing the fourth step of how the biosensor can be fabricated.
Fig. 18 is a diagram showing the fifth step of how the biosensor can be fabricated.
Fig. 19 is a diagram showing the sixth step of how the biosensor can be fabricated. Fig. 20 is a diagram showing how the surface of an embodiment of the present biosensor can be modified to prevent non-specific binding.
Fig. 21 is a diagram showing the final result on the biosensor surface of the surface modification process in Fig. 20.
Fig. 22 is a sectional diagram of an embodiment of the present invention of a biosensor system with a biosensor integrated with integrated circuit components.
Fig. 23 is a diagram of an embodiment of the present invention of a plurality of biosensors integrated with integrated circuit components.
Fig. 24 is a sectional diagram of a biosensor unit that, together with other similar units, makes up the plurality of biosensors integrated with the integrated circuit components in Fig. 24.
Fig. 25 is a diagram showing the first step of how an embodiment of a biosensor (sensor-chip) of the present invention can be fabricated by integrated circuit (IC) technology with a CMOS device integrated on the biosensor (sensor-chip) itself.
Fig. 26 is a diagram showing the second step of how the biosensor (sensor-chip) can be fabricated by integrated circuit (IC) technology.
Fig. 27 is a diagram showing the third step of how the biosensor (sensor-chip) can be fabricated by IC technology.
Fig. 28 is a diagram showing the fourth step of how the biosensor (sensor-chip) can be fabricated by IC technology. Fig. 29 is a diagram showing the fifth step of how the biosensor (sensor-chip) can be fabricated by IC technology.
Fig. 30 is a diagram showing the sixth step of how the biosensor (sensor-chip) can be fabricated by IC technology.
Fig. 31 is a diagram showing how an embodiment of a biosensor of the present invention can be use to detect ionic analytes (or molecules).
Fig. 32 is Cyclic Voltammetry (CV) (current vs. bias potential) graph taken by the sensor of Fig. 31. Fig. 33 is a diagram showing the first step of how an embodiment of a biosensor of the present invention can be used to detect macromolecules (e.g. DNS, RNA, protein).
Fig. 34 is a diagram showing the second step of the detection of the macromolecules. Fig. 35 is a diagram showing the third step of the detection of the macromolecules.
Detailed Description of the Preferred Embodiment
Although specific embodiments of the invention will now be described with reference to the drawings, it should be understood that various changes and modifications may be made without departing from the spirit, scope and contemplation of the invention. Indeed, the drawings and description herein are provided by way of examples, and not by way of limitations.
The first aspect of the invention relates to detection of various target analytes, especially ionic molecules (e.g. iron, chromium, lead, copper, calcium or potassium) and macromolecules (e.g. DNA, RNA or protein), using the principles of electrochemical detection. The principles of electrochemical detection require the use of a redox cell and an electrochemical reaction in the cell.
The redox cell is a device that converts chemical energy into electrical energy or vice versa when a chemical reaction occurs in the cell. Typically, the cell consists of three electrodes immersed into an aqueous solution (electrolyte) with electrode reactions occurring at the electrode-solution surfaces.
The cell consists of two electronically conducting phases (e.g., solid or liquid metals, semiconductors, etc.) connected by an ionically conducting phase (e.g. aqueous or nonaqueous solution, molten salt, ionically conducting solid). As an electrical current passes, it must change mode from electronic current to ionic current and back to electronic current. These changes of conduction mode are accompanied by reduction-oxidation reactions. Each mode changing reaction is called a half-cell.
Each electrochemical reaction is reduction-oxidation (redox) reaction that occurs in the redox cell. For example, in a spontaneous "chemical reaction" during the oxidation of hydrogen by oxygen to water, electrons are passed directly from the hydrogen to the oxygen. In contrast, in the spontaneous electrochemical reaction in the redox cell, two separate electrode reactions occur substantially simultaneously or in tandem.
An important feature of the redox cell is that the simultaneously occurring reduction-oxidation reactions are spatially separated. The hydrogen, for example, is oxidized at the anode electrode by transferring electrons to the anode electrode and the oxygen is reduced at the cathode electrode by accepting electrons from the cathode electrode. The overall electrochemical reaction is the sum of the two electrode reactions. The ions produced in the electrode reactions, in this case positive hydrogen ions and negative hydroxyl ions, will recombine in the solution to form the final product of the reaction: water.
During this process the electrons are conducted from the anode electrode to the cathode electrode through an outside electric circuit where the electronic current can be measured. The reaction can also be reversed; water can be decomposed into hydrogen and oxygen by the application of electrical power in an electrolytic cell.
A three-electrode system of the invention is an electrochemical cell containing a working electrode, a counter electrode (or auxiliary electrode), and a reference electrode. A current may flow between the working and counter electrodes, while the potential of the working electrode is measured against the reference electrode. This setup can be used in basic research to investigate the kinetics and mechanism of the electrode reaction occurring on the working electrode surface, or in electroanalytical applications. The detection module in the preferred embodiment in the instant invention is based of a three- electrode system.
The counter electrode is used to make an electrical connection to the electrolyte so that a current can be applied to the working electrode. The counter electrode is usually made of inert materials (noble metals or carbon/graphite) to avoid its dissolution. It has been observed in connection with the present invention that a small feature or small cross- section at the counter electrode will heat up the surrounding solution when a large current is pulled out from the counter electrode. Bubbles will be generated if the current is continuously overflowed, and ultimately dissolution of the electrode occurs. Thus, the bubble formation can be avoided by controlling the current and/or the electrode size. In a preferred embodiment of the present invention, the width of the counter electrode is large enough to avoid this heat up problem even at large current.
The reference electrode is used as a reference point against which the potential of other electrodes (typically that of the working electrode or measuring electrode) can be measured in an electrochemical cell. The few commonly used (and usually commercially available) electrode assemblies all have an electrode potential independent of the electrolyte used in the cell, such as a silver/silver-chloride electrode, calomel electrode, or hydrogen electrode. However, the present inventive entity has determined that a single layer electrode, such as a single layer of gold electrode, can fulfill the requirement for a reference electrode. In addition, other materials such as silver, copper, platinum, chromium, aluminum, titanium, nickel may also work as a single layer reference electrode under the right conditions.
The working electrode plays a central role in the electrochemical biosensors of the invention. The reaction occurring at the working electrode may be used to perform an electrochemical analysis of the electrolyte solution. It can serve either as an anode or a cathode, depending on the applied polarity. One of the electrodes in some "classical two- electrode" cells can also be considered a "working" ("measuring," "indicator," or "sensing") electrode, e.g., in a potentiometric electroanalytical setup where the potential of the measuring electrode (against a reference electrode) is a measure of the concentration of a species in the solution.
In a preferred three-electrode embodiment, the counter and reference electrodes are configured to extend generally about the periphering of the working electrode of the biosensor. Suitability configurations are seen in, for example, Figs. 3, 4, 5, 12, 13 and 23. In a preferred embodiment, the electrochemical biosensor is fabricated using microelectromechanical systems (MEMS) technology. Referring now to Fig. 3, a layer of silicon dioxide (SiO2, lOOOA) is deposited on a bare silicon wafer 200 (prime grade, p- type <100>, thickness 500-550. μm) and served as a pad layer underneath the silicon nitride (Si3N , lOOOA) to release stress and improve adhesion. A plurality of MEMS biosensors (such as biosensor 210) were fabricated with working electrodes of various dimensions (such as working electrode 220). Preferably each of the working electrodes is etched to form a well up to 350 μm in depth.
The nitride-coated silicon wafer 200 was patterned and bulk etched using KOH along the [111] and [100] crystal planes, and the depth of the well was controlled by KOH etching time and temperature. The 100. μm wide auxiliary (such as auxiliary electrode 230) and reference (such as reference electrode 240) electrodes are separated from their corresponding working electrode 220 by 200 μm. Fig. 3 shows a schematic of the pattern used in generating the MEMS biosensors (such as biosensor 210).
The nitride and oxide were removed by HF etching to release internal stress, and another oxide layer (5000A) was deposited for electrical isolation. Electrodes were patterned by PR5214 photo resist reverse imaging and lift-off process with electron beam deposition of Au(200θA)/Cr(20θA). Finally the wafer 200 was bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding Si areas. The hydrophobic nature of the surrounding area, along with the 3 -dimensional nature of the working electrode (such as working electrode 220), allows containment of a liquid droplet on the working electrode 220 during the initial sensing step of immobilizing to the working electrode molecules of interest in the solution. This design effectively minimized nonspecific binding of biomolecules to other areas of the MEMS array. Preferably the material used for all the electrochemical electrodes is gold (Au).
Several conducting materials, available in MEMS technology, were patterned on a silicon substrate by the lift-off process, and the characteristics of the three-electrode system were tested by cyclic voltammetry with ferricyanide solution. Different combinations of gold, platinum, titanium and aluminum electrodes were tested and the Au/Au/Au three-electrode system gave the best C-V curve and redox characteristics.
Fig. 4 shows an embodiment of biosensors fabricated on a square substrate 300 (such as silicon, gallium arsenide, plastic and/or glass) having a plurality of circular biosensors, such as biosensor 310. The biosensor 310 comprises a working electrode 320, a reference electrode 330, and a counter (auxiliary) electrode 340. As previously mentioned, preferably the electrodes are constructed out of a single layer of conducting materials, available in MEMS technology. Furthermore, all of the electrodes should preferably be constructed out of gold (Au).
Fig. 5 shows yet another preferred embodiment of the present invention. The biosensor 410 comprises a working electrode 420, a reference electrode 430 and a counter (auxiliary) electrode 440 fabricated on the substrate. Preferably, the electrodes are made out of gold. The working electrode 420 is formed in a built-in well 450 in the substrate up to 350 μm in depth. The well 450 is designed for confining a desired reagent within the well-defined space. As shown in Fig. 5, the well 450 fabricated by the microfabrication methods described herein, is bordered by (111) silicon planes after KOH etching. The working electrode 420, defined by the microfabrication methods described above, covers the entire well 450 surface. In operation, the above embodiments have the advantage of being easy to fabricate and can be fabricated together as a single series microfabrication process step on the substrate. However, successive microfabrication steps may also be employed for fabrication. Furthermore, as the following analysis and experiments below show, these low-cost and easy-to-fabricate biosensors are reusable and have the same robust and reversible electrochemical performance as conventional biosensors.
In one experiment, Cyclic Voltammetry (CV) analytical technique is used. CV is one of the most versatile analytical techniques used in the study of electroactive species and the characterization of biosensors. It is widely used as both an industrial and academic research tool in the fundamental characterization of electrochemical systems. In Cyclic Voltammetry, the potential is ramped from an initial potential (E0) to a maximum potential (Em) at a controlled (and typically fixed) sweep rate (V/sec). Fig. 6 illustrates this concept. Repeated cycles of reduction and oxidation of the analyte generate alternating anodic and cathodic currents in and out of the working electrode. Since the solution and/or reagent is not stirred, diffusion effects are observed at different analyte concentrations and different scan rates.
Separation of the anodic (ipa) and cathodic (ipc) current peaks can be used to predict the number of electrons involved in the redox reaction. The peak current is also directly proportional to the analyte concentration, C and scan rate, v. Experimental results are usually plotted as current versus potential, similar to the graph shown in Fig. 7.
In the CV scan shown in Fig. 7, the potential is graphed along the x-axis with more positive (or oxidizing) potentials plotted to the right, and more negative (or reducing) potentials to the left. The current is plotted on the y-axis with cathodic (i.e., reducing) currents plotted down along the negative direction, and anodic (i.e. oxidizing) currents plotted in the positive direction.
The analyte used in the following control experiment was potassium ferricyanide, K3Fe(CN)6 (329.26 g/mol), which contains an iron atom in the +3 oxidation state (Feιπ) in a buffer solution of potassium nitrate, KNO (101.11 g/mol). At the surface of a working electrode, a single electron can be added to the ferricyanide anion. This will cause it to be reduced to the ferricyanide anion, Fe"(CN)6 4', which contains an iron atom in the +2 oxidation state (Fe11). This simple, one electron exchange between the analyte and the electrode is a well behaved, reversible reaction. This means that the analyte can be easily reduced to Feπ(CN)6 4" and then easily oxidized back to Feπι(CN)6 3".
A redox couple is a pair of analytes differing only in oxidation state. The electrochemical half-reaction for the Fem(CN)6 3" / Fe"(CN)6 4" redox couple can be written as follows: Fe1" (CN)6 3~ + e' <= Fe" (CN)6 4~ E0 = +0.358 V NHE). (1) The voltammogram shown in Fig. 8 exhibits two asymmetric peaks, one cathodic
(ipC) and the other anodic (ipa). Using a standard reference electrode, such as the normal hydrogen electrode (NHE), the formal potential associated with this half-reaction is near +358 mV. If the working electrode is held at a potential more positive than +400 mV, then the analyte tends to be oxidized to the Fe'"(CN)6 3~ form. This oxidation at the working electrode causes electrons to go into the electrode from the solution resulting in an anodic current. At potentials more negative than +400 mV, the analyte tends to be reduced to Fe'" (CN)6 ~ . This reduction at the working electrode causes electrons to flow out of the electrode into the solution resulting in a cathodic current. Since the preferred sensor design embodiment of the present invention does not utilize a standard reference electrode like NHE, silver/silver chloride (Ag/AgCl) or saturated calomel electrode (SCE), and since all three electrodes are gold (Au), the rest (unbiased) potential in this experiment is close to zero volts.
The important parameters of a cyclic voltammogram are the magnitudes of the cathodic and anodic peak currents (ipc and ipa, respectively) and the potentials at which these currents are observed (Epc and Epa, respectively). Using these parameters, it is possible to calculate the formal reduction potential (E0 - which is centered between Epa and Epc ) and the number of electrons (n) transferred in the charge transfer reaction. The peak current (ipa or ipc) can be expressed by the Randles-Sevcik equation:
Figure imgf000014_0001
where, n = number of electrons appearing in half-reaction for the redox couple F= Faraday's constant (96,485 C/ mol) A = electrode area (cm2) v = rate at which the potential is swept (V/sec) D = analyte 's diffusion coefficient (cm2/sec) R = universal gas constant (8.314 J / mol K) T - absolute temperature (K) At 25°C, the Randles-Sevcik equation can be reduced to the following:
ip = (2.687xl0s)n3/2vx/2D1/2AC (3)
where the constant has units (i.e., 2.687x105 C mol"1 V~1/2).
The Randles-Sevcik equation predicts that the peak current should be proportional to the square root of the sweep rate when voltammograms are taken at different scan rates. As shown in Fig. 9, the plot of peak current versus the square root of sweep rate yields a straight line. The Randles-Sevcik equation can be modified to give an expression for the slope of this straight line as follows,
Slope = (2.687 xIO'CmoF -^n^D^AC (4)
The scan rate dependence of the peak potentials and peak currents are used to evaluate the number of electrons participating in the redox reaction as well as provide a qualitative account of the degree of reversibility in the overall reaction. The number of electrons transferred in the electrode reaction («) for a reversible redox couple is determined from the separation between the peak potentials (AEp = Epa - Epc). For a simple, reversible
(fast) redox couple, the ratio of the anodic and cathodic peak currents should be equal to one. The results of the experiment using the preferred sensor design deviate only slightly from unity. Large deviations would indicate interfering chemical reactions coupled to the electrode processes, but slight deviations from unity merely suggest a non-ideal system.
Fig. 10 shows the CV scan for 12 consecutive cycles with minor deviations from the first cycle. These results clearly indicate a highly reversible system and robust cell design. Besides being easy and cheap to fabricate while having the same performance as the conventional sensor, the above embodiments of the present invention also have the advantages of being compatible with the integrated circuit (IC) and the MEMS fabrication process. Furthermore, as shown in Figs. 3 and 4 these embodiments are capable of being constructed in an array of biosensors and in a small area.
A second aspect of the present invention relates to confinement of a reagent and/or solution in a biosensor using surface tension at small scale. The reagent and/or solution contains the target analyte(s) and/or the chemical(s) needed for biosensing.
A biosensor's performance is mainly determined by its specificity and sensitivity. The concept of confining the reagent on a substrate, such as silicon, was discovered in investigations relating to the present invention to be a solution for reducing the high level of detection noise caused by the non-specific binding of the analyte or other reagent or solution components, such as Horseradish Peroxidase (HRP), onto regions arund the periphery of the working electrode and causing high detection noises and creating high false positive rates. HRP is used in the instant embodiment as a signaling enzyme. To verify that noise does come from the HRP residual at the surface, a simple test was done to estimate the contribution of this unwanted binding. HRP was introduced to a bare silicon chip, followed with several wash steps before the addition of the substrate solution. A very high level of enzymatic reaction was observed immediately after adding the substrate solution.
As expected, HRP, like other proteins, sticks to the silicon surface easily and tightly. Several commercial wash solutions and blocking protein were tested without any significant improvement of reducing non-specific binding. Thus, it was determined that the best way to avoid the effects ofundesirable binding is to prevent it from happening in the first place. The area outside the working electrode of the biosensor of the invention need not encounter the HRP solution in order to achieve biological sensing. In the prior art, however, it is conventional to immerse all three electrodes in aqueous system all the time and all the reagents would flow through all the surface area.
The simplest way, discovered by the present inventive entity, to confine certain reagents within a well-defined space is to form a well in the biosenser. As shown in Fig. 5, a micro fabricated well is bordered by silicon crystal planes 400 after KOH etching. The working electrode 420 defined by a lift-off process covers the whole well surface. Thus, a reagent or solution containing components capable of non-specific binding (e.g. HRP) may initially be confined to the well area to achieve the desired binding to the electrode in the well (preferably the working electrode), while avoiding non-specific binding to the periphery region of the biosensor. The reagent or solution can then be washed off the biosensor and additional reagents and/or solutions later added to complete the sensing process.
The importance of surface and material science cannot be underestimated when designing a biosensor. An unacceptable amount of non-specific binding may still occur during the washing process while the diluted reagent is flowing around the wafer surface. The time for the wash solution containing HRP, for example, to stay on the periphery region is much longer than the binding time constant. Therefore, besides fabricating the well structure, the surface of the periphery area preferably should be protected by other mechanisms.
Thus, another way to keep the surface from contacting undesired reagents or binding components is to make it hydrophobic. Silanation of silicon surface is widely used to prevent suspended structures from sticking to substrate by surface tension. This approach was used here to prevent the direct contact of biomolecules to the periphery area of the silicon substrate.
Silane-based molecules with various functional terminal groups have been used to modify surface properties of silicon wafers, silicon nitride chips, and atomic force microscopy (AFM) tips. Silane compounds for the surface modification form robust monolayers chemically tethered to silicon oxide surfaces as a result of hydrolysis of terminal Si(Cl)n or SiO-C2H5 groups.
The performance of artificial materials in contact with biological systems is determined by the surface interactions of the two materials. Since the surface interaction could result in noise increasing and structure damage, surface modification is one way to avoid this problem. The easiest way is to change the surface property of the structure or by using plastic. By converting the surface into a biocompatible or bio-inert surface, nonspecific interaction will be limited to a minimum or eliminated by the fabrication of a molecular layer firmly tethered to the surface. The most widely implemented approach uses a self-assembly monolayer (SAM) from thiol molecules by chemisorption onto a gold surface and silane molecules to form SAM on a silicon oxide surface according to well- established procedures developed in the 80's mostly by C. D. Bain, and G. M. Whitesides. Other materials such as sol gel and carbon paste may also be used to modify the surface.
In operation, referring now to Fig. 11(a), the reagent is shaped by the hydrophilic working electrode and a droplet is nicely formed over the Au working electrode surface. Fig. 11(b) demonstrates that with increasing volume of the reagent, the area covered by the reagent will gradually expand and then cover the other two electrodes. Fig. 11(c) shows that when an excess stream of reagent is administered by pipette, a ball 465 of reagent will be formed instead of spreading out on the hydrophobic silicon substrate.
Turning now to Figs. 12 and 13, each individual electrode will be in contact with electrolyte and/or analyte and other components 509 only when needed by using controllable surface property and surface tension force at small scale. Referring now only to Fig. 13, for electrochemical detection, all electrodes have to be immersed in the electrolyte and/or analyte 509 only at detection time. For most applications, the majority of time is spent on sample preparation as depicted in Fig. 12 to immobilize the electrolyte and or analyte 509 onto working electrode 510. In this embodiment as shown in Figs. 12 and 13, the coverage of the electrolyte and/or analyte 509 over the electrodes is controlled by surface property and reagent volume 520. Thus, the above embodiments take advantage of surface tension at small scale to confine the electrolytes and or analytes and other components. In addition, the above embodiments require much less reagent (in the order of pL to mL); and eliminate the need for bulk solution. Furthermore, because of the small amount of reagent use, the above embodiments have the advantages of having the analyte close to the electrodes (pm to mm), whereby adequate exposure of analyte to the electrodes can be achieved using diffusion effects alone, and the need for stirring or other mixing is eliminated. Another advantage of the above embodiments is the ease of the control and/or change of the coverage of electrolyte and/or analyte over the electrodes. Moreover, the above reagent and/or solution confinement embodiments reduce the loss of target analytes and improve the sensitivity of the biosensor by greatly reduce non-specific binding. In addition, the above embodiments are relatively immune to shaking of the biosensor and are suitable for portable or handheld systems. Lastly, due to the surface tension confinement effect, the reagent will be held firmly by surface tension even the sensor is flipped upside down.
Optionally, as shown in Figs. 12 and 13, the biosensor embodiments can be equipped with a reaction well 530 to help hold the reagent 520 in place and to control the shape of the reagent 520. However, such a well 530 is not required and the confinement of the reagent 520 can be achieved by a simple flat surface.
Referring now to Fig. 14, this shows the first step of a preferred fabrication of an embodiment of the present biosensor (sensor-chip). As shown in Fig. 14, a layer of silicon dioxide (SiO2, lOOOA) 620 is first deposited on a bare Si wafer 610 (prime grade or test grade, p-type or n-type <100>, thickness 500-550 μm). The silicon dioxide 620 serves as a pad layer underneath the silicon nitride 630 (Si3N4, lOOOA) to release stress and improve adhesion.
Next, referring to Figs. 15 and 16, the nitride wafer 600 is patterned (Fig. 15) by mask 640 and bulk etched using KOH along the [111] and [100] crystal planes (Fig. 16). The etching creates a well 650 and the depth of the well 650 (up to 350 μm) is controlled by KOH etching time and temperature.
Referring now to Fig. 17, the nitride 630 and oxide 620 were then removed by HF etching to release internal stress, and another oxide 660 layer (5000A) is deposited for electrical isolation.
Figs. 18 and 19 show a lift-off process for the fabrication of the necessary electrodes. The electrodes for biosensing are patterned on substrate 610 by using PR5214 photo resist layer 670 on silicon dioxide layer 660. Next, a mask 671 is used to transfer a desired pattern onto photo resist layer 670 by using reverse imaging process (which includes the removal unwanted photo resist layer 670). A layer of gold Au (2000A) 680 is electron-beam deposited on silicon dioxide layer 660 with the desired photo resist pattern layer 670. Preferably, a deposition of an adhesive 690, such as chromium (Cr 20θA) occurs before the deposition of Au(200θA) 680 to improve the adhesion of the Au onto the silicon dioxide layer 660. In addition, other materials such as titanium or glue may also work as an adhesive. Lastly, referring to Fig. 19, any photo resist 670 and unwanted Au 680 and Cr 690 are removed by dissolving the photo resist pattern layer 670. Finally the wafer 600 is bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding silicon areas.
Preferably, the surface of the biosensor (sensor chip) is modified by a surface modification step to prevent non-specific binding. As an example, referring to Fig. 20, a macromolecule biosensor's surface can be modified by the steps of first cleaning the Au surfaces 680 with concentrated "Piranha" solution (70 vol% H2SO4, 30 vol% H2O2) and thoroughly rinsed with deionized water (dH2O). Next, referring to Fig. 20, surface modification material such as SAM of biotin-DAD-C12-SH (12-mercapto(8-biotinamide- 3,6-dioxaoctyl)dodecanamide) Roche GmBH, Germany is deposited. For depositing a SAM of biotin-DAD-C12-SH), the procedure of Spinke, et al. (1993) was used wherein samples were incubated for ~18 hours in a 50mM solution of biotin-DAD-C12-SH in ethanol with 4.5xl0-4M 11-mercapto-l-undecanol (Aldrich Chemical Co., 44,752-8) and rinsed with ethanol and water. Finally, referring to Fig. 21, the biotin-coated Au surfaces 700 were then exposed to a 1.0 mg/ml streptavidin solution for ~10 minutes and rinsed again with dH2O to form a streptavidin-coated Au surface 710.
A third aspect of the present invention relates to the integration of an entire electrochemical (redox) biosensor onto a single integrated circuit (IC) chip. An integrated circuit (IC) is defined as a tiny wafer of substrate material upon which is etched or imprinted a complex of electronic components and their interconnections. MEMS devices have distinctive properties as a result of small features but the signal level from MEMS-based sensor is relatively low compared to a conventional sensor. Sensitivity can be improved by using an off-chip amplification module but it also increases the noise and the system size. Thus, the present inventive entity designed an on- chip amplification circuitry (amplification circuit incorporated on the biosensor chip itself) and detection circuitry (such as providing bias potential, current measurement, sequential control and signal processing) to reduce the inter-chip interference. Both the detection circuitry and the amplification circuitry can be provided by using bipolar transistor (BJT) and/or complementary metal-oxide-semiconductor (CMOS) devices.
In a present embodiment, an on-chip amplification device is equipped underneath the working electrode, which is typically the largest area of the electrochemical sensor cell. Analogous to the open-base bipolar (BJT) photosensor, the base region receives the current from the transducer with the current gain beta β, which ranges from 80-150. There are two types of BJT which can be implemented with an electrochemical cell, vertical BJT and horizontal BJT. The current gain is determined by the length of base region. In vertical BJT, this is a function of ion implementation energy and doping concentration. In horizontal BJT, the length is a function of lithography resolution, ion implementation angle and thermal diffusion. For these reasons the vertical BJT is more reliable in terms of chip-to-chip or in-chip gain uniformity.
Fig. 22 shows in cross-section an embodiment of in-chip amplifier circuitry that preferably includes a biosensor comprising three metal electrodes used for electrochemical sensing. Preferably, the three electrodes are fabricated using integrated circuit (IC) processes and have a relatively large area for interconnection and isolation. The entire biosensor chip 700 can be stacked in two stages (electrochemical sensing stage 730 and bipolar transistor (BJT) amplifying stage 720) and the Au working electrode 710 can act as an electromagnetic shield for the BJT device 720. The working electrode contacts (such as contacts 733, 734, 735, 736, 737 and 738) make contact to a contact interface 740 and also increase the surface area of the working electrode 710 for higher signal. All contact and electrode structures can be made with a single layer of metal such as gold. In addition, biosensor chip 700 comprises a silicon substrate 744. Substrate 744 includes a base region 743, a collector 742 and an emitter 741. Base region 743 receives electron flux from working electrode 710. Collector 742 is connected to a power source and provides the current gain under certain base conditions for amplification reasons. A resulting current from base 743 and collector 742 is then measured at emitter 741.
Referring still to Fig. 22, metal interconnect 750 is connected to signal output (not shown) and emitter 741. Gold working electrode 710 connects to base region 743 through a first layer 760 of silicon dioxide via etching. A second layer of silicon dioxide 770 isolates the BJT 720 and signal line while first layer 760 isolates the three electrodes (working electrode 710, reference electrode 780, and counter electrode 790). The contacts (such as contacts 733, 734, 735, 736, 737 and 738) on working electrode 710 increase the surface area and also form a solid contact with BJT 720. The dimension of the contacts (about a few tenths μm each) is much larger than the size of the optional protein SAM (about a few tenths A) on working electrode 710 so the protein adsorption will not be affected. Fig. 24 shows a sectional view of another preferred embodiment of a biosensor unit 800 representing a preferred embodiment of a plurality of biosensors on a substrate
805 shown in Fig. 23. As seen in Fig. 24, the biosensor unit 800 comprises a CMOS and/or BJT layer 830, an electrochemical layer 840 and a reagent containment layer 850, and is generally similar to the Fig. 22 embodiment already described.
Thus, as shown in Figs. 22, 23 and 24, the third aspect of the present invention is related to a biosensing system that allows for direct integration without chip-to-chip connection of the components in the sensor with other components needed for biological detection. The system does not require an external component (instrument) or requires only a minimum number of external components to constitute a complete system. Furthermore, this all-in-one biosensing system reduces the cost and noise level of biological sensing and simplifies the process of such sensing.
Referring now to Fig. 25, which shows an example of how an embodiment of the present biosensor (sensor-chip) can preferably be fabricated by integrated circuit (IC) technology with a CMOS device and/or devices. In addition, a BJT device and/or devices (not shown) may also be included. As shown in Fig. 25, an integrated circuit (IC) is fabricated on semiconductor substrate 900 with a layer of silicon dioxide 905 with a poly silicon gate 907 on top of a active well region 909. Poly silicon gate 907 is fabricated from an amorphous silicon and it is used for switching the transistor comprises low resistance source 917 and drain 919.
Referring now to Fig. 26, an oxide layer (5000A) 920 is next deposited on the substrate 900 for electrical isolation. Referring now to Fig. 27, the silicon dioxide layer 920 is then selectively etched by using lithography and etching methods to form an electric connection hole 940. Referring now to Fig. 28, a conducting plug 950 is then applied into the contact hole 940. Conducting plug 950 is used as interconnection between conductive electrode 960 with low resistance source 917 and/or drain 919.
Fig. 29 shows a lift-off process for the fabrication of the necessary electrodes. The electrodes for biosensing are patterned on substrate 900 by using PR5214 photo resist layer 970 on silicon dioxide layer 920. Next, a mask 921 is used to transfer a desired pattern onto photo resist layer 970 by using reverse imaging process (which includes the removal of unwanted photo resist layer 970). A layer 960 of gold Au(200θA) and/or Cr(20θA) is electron-beam deposited on silicon dioxide layer 920 with the desired photo resist pattern layer 970. Lastly, referring to Fig. 30, any photo resist 970 and unwanted Au/Cr 960 are removed by dissolving the photo resist pattern layer 970. Referring now to Fig. 31, this shows an example of how an embodiment of the present biosensor (sensor-chip) 1000 can preferably be used to detect an ionic molecule, such as iron (Fe). In this embodiment, the sensor surface 1010 is not modified and is ready to use after the post fabrication cleaning as shown in the above fabrication embodiment. The analyte used in this embodiment was potassium ferricyanide, K3Fe(CN)6 (329.26 g/mol), which contains an iron atom in the +3 oxidation state (Fe111) in a buffer solution of potassium nitrate, KNO3 (101.11 g/mol). As shown, in Fig. 31, the mixed solution 1020 with analytes (or reagent with electrolytes) is applied onto all three electrodes (working 1030, reference 1040 and counter electrode 1050). The volume of the solution 1020 is adjusted so the droplet is confined over all three electrodes by surface tension forces.
Next, referring to Fig. 32, the Cyclic Voltammetry (CV) current vs. bias potential was measured using a CH Instruments 660A Electrochemical Workstation with a picoamp booster and faraday cage. The potential is swept between 0.1 volt and -0.4 volt and the current is measured through working electrode 1030.
After the measurement, the Au surfaces were cleaned with acetone, alcohol, and concentrated "Piranha" solution (70 vol% H2SO , 30 vol% H2O2) and thoroughly rinsed with deionized water (dH2O) if the sensor 1000 is to be re-used.
Fig. 33 shows a preferred embodiment for macromolecules (e.g., DNA, RNA, protein) detection. Preferably, the detection of the concentration of macromolecules (DNA, RNA, protein) does include the sensor surface modification of biotin/Streptavidin layer. The sensor surface is modified with a proper biochemical solution to form a streptavidin layer after the post-fabrication cleaning as shown in the surface modification process described above. After the surface of the biosensor 1100 has been modified, the amperometric biosensing of pathogen is conducted by first adding 50 ml of lysis reagent (0.4 M NaOH) to a 250 ml sample of bacteria solution and incubated for 5 minute at room temperature. 100 ml of probe solution (anchoring and signaling probes) was then added, and the mixture was incubated for 10 min. at 65°C. 5 μL of the lysed E. coli/probe solution mixture 1120 is then placed on the streptavidin coated working electrode 1130 of the biosensor 1100 and incubated for 10 min. at room temperature.
Referring now to Fig. 34, the biosensor 1100 is then washed with biotin wash solution (Kirkegaard and Perry Laboratories, 50-63-06). Next, 5 μL 1140 of Anti-Fl-POD (Anti-fluorescein peroxidase, 150U, Roche Inc., 1 426 346), diluted to 0.75U/ml or 0.15 U/ml with dilutant (PBS/0.5%Casein) is placed on the working electrode 1130 and incubated for 10 minutes at room temperature.
Referring now to Fig. 35, the biosensor is then washed again with wash solution. After the wash, 10 μL 1160 of K-blue substrate (Neogen Corp., 300176) is placed on the biosensor in such a way that all three electrodes (working 1130, reference 1140, counter 1150) are covered by the substrate solution 1160. Finally, the electrochemical (more specifically the redox) measurements are immediately taken. Amperometric current vs. time is measured using a CH Instruments 660A Electrochemical Workstation with a picoamp booster and faraday cage. Samples on the biosensor are to be measured sequentially. The voltage should be fixed at -0.1V (vs. reference), and a cathodic current (amperometric signal) reading should be taken in 20 seconds because at 20 seconds, the current values should reach steady-state. Cell concentration (cell number) is determined by using serial dilutions and culture plate counting.
After the measurement, the Au surfaces should then be cleaned with acetone, alcohol, and concentrated "Piranha" solution (70 vol% H2SO4, 30 vol% H2O2) and thoroughly rinsed with deionized water (dH2O) if the sensor 1100 is to be re-used.
Preferably, in any of the above biosensor embodiments the biosensor is calibrated before actual sample detection. The biosensor is preferably calibrated with a calibrating solution that contain a known amount of target analyte(s) and another calibrating solution that contains an undetectable amount (e.g., none) of the target analyte(s). Optionally, the biosensor may be calibrated with a plurality of calibrating solutions. Each of the plurality of calibrating solutions respectively contains a known amount of the target analyte. Preferably, the calibrating solutions contain the target analytes at different level of concentrations. These calibrations solutions are measured on the biosensor by the above described detection process to obtain a reference signal and/or reference signals. The reference signal and/or signals are then compared with the measured signal from a sample solution to determine the presence and quantity of the analyte in the sample reagent. The determination of the presence and quantity (e.g., concentration or absolute amount) of the analyte(s) can be determined by conventional biological interpolation methods. Appendix A provides additional material to further characterize the present invention.
All references herein to outside literature, including references made within the appendix of this application, are hereby incorporated by reference to the extent that they supplement, explain, provide a background for or teach methodology, techniques and/or compositions employed herein. In addition, the references listed below are also incorporated herein by reference to the extent that they supplement, explain, provide a background for or teach methodology, techniques and/or compositions employed herein.
References:
"USAF PAMPHLET ON THE MEDICAL DEFENSE AGAINST BIOLOGICAL MATERIAL - Medical Defense Against Biological Material" Defense Technical Information Center, released on 11 Feb 1997 Abbott, N.L.; Gorman, C.B.; Whitesides, G.M. "Active control of wetting using applied electrical potentials and self-assembled monolayers" Langmuir , 11, Issue 1, January 1995, 16-18
Abbott, N .; Rolison, D.R.: Whitesides, G.M., "Combining micromachining and molecular self-assembly to fabricate microelectrodes" Langmuir, 10, Issue 8, August 1994, 2672-2682
Abdel-Hamid, I. Ivnitski, D., Atanasov, P., Wilkins, E., 1998. Fast Amperometric Assay for E. coli 0157:H7 using partially immersed immunoelectrodes. Electroanalysis 10 (11), 758-763. Abdel-Hamid, I., Ivnitski, D., Atanasov, P., Wilkins, E., 1999. Flow-through immunofiltration assay system for rapid detection of E. coli 0157:H7. Biosens. Bioelect. 14, 309-316.
Andrade, J.D., "Surface and Interface Aspects of Biomedical Polymers: Protein Adsorption" Plenum, New York, 1985 Bain, C. D.; Troughton, E. B.; Tao, Y.T.;Evall, J.; Whitesides, G. M.; Nuzzo, R.
G., J. Am. Chem. Soc. 1989, III, 321
Baxter, Ian; Cother, Lisa D.; Dupuy, Carole; Lickiss, Paul D.; White, Andrew J.P.; and Williams, David J. "Hydrogen Bonding to Silanols" Department of Chemistry, Imperial College of Science, Technology and Medicine, London S W7 2AY, UK Beam, K.E. "Anisotropic Etching of Silicon," IEEE Transactions on Electron
Devices, October, 1978
Becker, E.W.; Betz, H.; Ehrfeld, W.; Glashauser, W.; Heuberger, A.; Michel, H.J.; Munchmeyer, D.; Pongratz, S.; and Siemens, R.V. "Production of Separation Nozzle Systems for Uranium Enrichment by a Combination of X-Ray Lithography and Galvano- plastics," Naturwissenschaften, 69, (1982), 520 to 523.
Blake, C, Gould, B.J., 1984. Use of enzymes in immunoassay techniques. A review. Analyst 109, 533-547.
Brody, J.P.; Yager, P.; Goldstein, R.E. and Austin, R.H. "Biotechnology at low Reynolds numbers" J. Biophys. 71, 1996, 3430-3441 Chen, Y.F., Yang, J.M., Gau, J.J., Ho, CM., Tai, Y.C., 2000. Microfluidic System for Biological Agent Detection. Proceedings of the 3rd International conference on the interaction of art and fluid mechanics, Zurich, Switzerland.
Chen, Y.F.; Yang, J.M.; Gau, J.J.; Ho, CM.; and Tai, Y.C., "Microfluidic System for Biological Agent Detection" The 3rd International conference on the interaction of Art and Fluid Mechanics, Zurich, Switzerland , 2000.
Cooper, N. "The Human Genome Project," Los Alamos Lab, Science, 20,1-338. 1992. Cussler, E. L., Diffusion - mass transfer in fluid systems, 2nd edition, , Cambridge University Press, 1997
Darst, S.A.; Ahlers, M.; Meller, P.H.; Kubalek, E.W.; Blankenburg, R.; Ribi, H.O.; Ringsdorf, H.; Kornberg, R., "Two-Dimensional Crystals of Streptavidin on Biotinylated Lipid Layers," Biophys J. 59 ( 1991 ) 387-396.
Dunford, H.B., 1991. Horseradish peroxidase: structure and kinetic properties. In: Everse, j., Everse, K.E., Grisham, M.B. (Eds.), Peroxidases in Chemistry and Biology, vol. 2. CRC Press, Boca Raton, FL, pp. 1-24.
Ehteshami, G.; Rana, N.; Raghu, P.; and Shadman, F. "Interactions of impurities with Silicon and Silicon dioxide during oxidation", Center for Micro-contamination Control, NSF I/U CRC
Feynman, Richard P. "There's Plenty of Room at the Bottom" Engineering and Science, Caltech, February 1960
Gau, J.J., Lan, E. H., Dunn, B., Ho, CM., 2000. Enzyme-based electrochemical biosensor with DNA array chip. Proceedings of the fourth International Symposium on Micro Total Analysis Systems (μTAS), Enschede, The Netherlands.
Ghindilis, A.L., Atanasov, P., Wilkins, E., 1997. Enzyme-catalyzed direct electron transfer: Fundamentals and analytical applications. Electroanalysis 9, 661-674.
Gorton, L., Lindgren, A., Larsson, T., Munteanu, F.D., Ruzgas, T., Gazaryan, I., 1999. Direct electron transfer between heme-containing enzymes and electrodes as basis for third generation biosensors. Anal. Chim. Acta 400, 91-108.
Hall, E.A.H., 1991. Biosensors. Prentice Hall, Englewood Cliffs, New Jersey.
Haussling, L., Ringsdorf, H., Schmitt, F.J., Knoll, W., 1991. Biotin-functionalized self-assembled monolayers on gold: surface plasmon optical studies of specific recognition reactions. Langmuir 7 (9), 1837-1840.
Ho, CM., Tai, Y.C., 1996. Review: MEMS and its applications for flow control. J. Fluids Eng. 118, 437-447.
Ho, CM., Tai, Y.C, 1998. Micro-electro-mechanical systems (MEMS) and fluid flows. Ann. Rev. Fluid Mech. 30, 579-612. Hou, S.F., Yang, K.S., Fang, H.Q., Chen, H.Y., 1998. Amperometric glucose enzyme electrode by immobilizing glucose oxidase in multilayers on self-assembled monolayers surface. Talanta 47, 561-567.
Howe, R.T. and Muller, R.S. "Polycrystalline Silicon Micromechanical Beams," Journal of the Electrochemical Society: Solid State Science and Technology, June, 1983. Ivnitski, D., Abdel-Hamid, I., Atanasov, P., Wilkins, E., 1999. Biosensors for detection of pathogenic bacteria. Biosens. Bioelect. 14, 599-624. Ivnitski, D., Abdel-Hamid, I., Atanasov, P., Wilkins, E., Strieker, S., 2000. Application of Electrochemical Biosensors for Detection of Food Pathogenic Bacteria. Electroanalysis 12 (5), 317-325.
Jung, L.S., Nelson, K.E., Campbell, C.T., Stayton, P.S., Yee, S.S., Perez-Luna, V., Lopez, G.P., 1999. Surface plasmon resonance measurement of binding and dissociation of wild-type and mutant streptavidin on mixed biotin-containing alkylthiolate monolayers. Sensors Actuators B 54, 137-144.
Kane, R.S., Takayama, S., Ostuni, E., Ingber, D.E., Whitesides, G.M., 1999. Patterning proteins and cells using soft lithography. Biomaterials 20, 2363-2376. Kevles, D., and L. Hood . "The Code of Codes: Scientific and Social Issues in the
Human Genome Project", Harvard University Press, Cambridge, Mass., 1992.
Lahiri, J., Ostuni, E., Whitesides, G.M., 1999. Patterning Ligands on Reactive SAMs by Microcontact Printing. Langmuir 15 (6), 2055-2060.
Lindgren, A., Munteanu, F.-D., Gazaryan, I.G., Ruzgas, T., Gorton, L., 1998. Comparison of rotating disk and wall-jet electrode systems for studying the kinetics of direct and mediated electron transfer for horseradish peroxidase on a graphite electrode. J. Electroanal. Chem. 458, 113-120.
Marrazza, G.; Chianella, I.; Mascini, M., "Disposable DNA Electrochemical Biosensors for Environmental Monitoring", Analytica Chimica Acta 387 (1999), 297-307. Motesharei, K., Myles, D.C., 1998. Molecular recognition on functionalized self- assembled monolayers of alkanethiols on gold. J. Am. Chem. Soc. 120 (29), 7328-7336.
Murthy, A.S.N, Sharma, J., 1998. Glucose oxidase bound to self-assembled monolayers of bis(4-pyridyl) disulfide at a gold electrode: Amperometric determination of glucose. Anal. Chim. Acta 363, 215-220. Nathanson, H.C; Newell, W.E.; Wickstrom, R.A.; and Davis, J.R., Jr., "The
Resonant Gate Transistor," IEEE Transactions on Electron Devices, March, 1967.
Ooka, A. A., Kuhar, K.A., Cho, N., Garrell, G.L., 1999. Surface Interactions of a Homologous Series of a,w-Amino Acids on Colloidal Silver and Gold. Biospectroscopy 5, 9-17. Ostuni, E., Yan, L., Whitesides, G.M., 1999. The interaction of proteins and cells with self-assembled monolayers of alkanethiolates on gold and silver. Colloids Surfaces B 15, 3-30.
Rao, J., Yan, L., Xu, B., and Whitesides, G.M., 1999. Using surface plasmon resonance to study the binding of vancomycin and its dimer to self-assembled monolayers presenting D-Ala-D-Ala. J. Am. Chem. Soc. 121 (11), 2029-2030. Revell, D.J., Knight, J.R., Blyth, D.J., Haines, A.H., Russell, D.A., 1998. Self- assembled carbohydrate monolayers: formation and surface selective molecular recognition. Langmuir 14 (16) 4517-4524.
Ruzgas, T., Gorton, L., Emne'us, J., Marko-Varga, G., 1995. Kinetic models of horseradish peroxidase action on a graphite electrode. J. Electroanal. Chem. 391, 41-49.
Ruzgas, T.; Csδregi, E.; Emneus, J.; Gorton, L.; Marko-Varga, G., "Peroxidase- modified electrodes: Fundamentals and application", Analytica Chimica Acta 330 (1996), 123-138.
Sieval, A.B.; Demirel, A.L.; Nissink, J.W.M.; Linford, M.R.; van der Maas, J.H.; de Jeu, W.H.; Zuilhof, H. and Sudhδlter, E.J.R. "Highly stable Si-C linked functionalized monolayers on the silicon(lOO) surface." Langmuir 1998, 14, 1759-1768.
Sieval, A.B.; Zuilhof, H.; Sudhδlter, E.J.R.; Schuurmans, F.M.; and Sinke, W.C "Surface passivation of silicon by organic monolayers." Proceedings of the 2nd World Conference and Exhibition on Photovoltaic Solar Energy Conversion, Vienna, 1998 Sigal, G.B., Bamdad, C, Barberis, A., Strominger, J., and Whitesides, G.M., 1996.
A self-assembled monolayer for the binding and study of histidine-tagged proteins by surface plasmon resonance. Anal. Chem. 68, 490-497.
Spinke, J., Liley, M., Guder, H.J., Angermaier, L., Knoll, W., 1993. Molecular recognition at self- assembled monolayers: the construction of multicomponent multilayers. Langmuir 9 (7), 1821 - 1825.
Sun, X., He, P., Liu, S., Ye, Jiannog, Fang, Y., 1998. Immobilization of single- stranded deoxyribonucleic acid on gold electrode with self-assembled aminoethanethiol monolayer for DNA electrochemical sensor applications. Talanta 47, 487-495.
Taylor, Robert "Bioterrorism Special Report: All fall down", New Scientist, Volume 150, Issue 2029, 11 May 1996
Trimmer, W.S.N. "Microrobots and Micromechanical Systems" Sensors and Actuators, 19, Number 3, September 1989, 267 - 287
Trimmer, William "Grand in Purpose, Insignificant in Size" The tenth Annual International Workshop on MEMS, Nagoya, Japan, Proceedings IEEE Catalog Number 97CH36021, 1997, 9 - 13.
Ulman, A. Ultrathin Organic Films from Langmuir Blodgett to Self-Assembly; Academic Press: New York, 1991.
Ulman, A., An Introduction to Ultrathin Organic Films From Langmuir Blodgett to Self-Assembly, Academic Press, Inc., San Diego, CA, 1991 Ulman, A., Ed. Characterization of Organic Thin Films; Butterworth-Heinemann:
Boston, 1995.
Voet, Donald; Voet, J. " Biochemistry, 2nd edition" Wiley, New York, 916-919 Wagner, P.; Hegner, M.; Guntherodt, H-J; Semenza, G., "Formation and in Situ Modification of Monolayers Chemisorbed on Ultraflat Template-Stripped Gold Surfaces," Langmuir 11 (1995) 3867-75.
Wang, J., Rivas, G., Cai, X., Palecek, E., Nielsen, P., Shiraishi, H., Dontha, N., Luo, D., Parrado, C, Chicharro, M., Farias, P.A.M, Valera, F.S., Grant, D.H., Ozsoz, M., Flair, M.N., 1997. DNA electrochemical biosensors for environmental monitoring. A Review. Anal. Chim. Acta 347, 1-8.
Wang, T.H., Chen Y.F., Masset S., Ho, CM., Tai, Y.C., 2000. Molecular beacon based micro biological detection system. Proceedings of the 2000 International Conference on Mathematics and Engineering Techniques in Medicine and Biological Sciences (METMBS'2000) Las Vegas, Nevada.
Weber, P.C, Ohlendorf, D.H., Wendoloski, J.J., Salemme, F.R., 1989. Structural Origins of High- Affinity Biotin Binding to Streptavidin. Science 243, 85-88.
Whitesides, G. M.; Laibinis, P. E., Langmuir 1990, 6, 87. Wink, Th.; van Zuilen, S.J.; Bult, A.; and van Bennekom, W.P., "Self-assembled
Monolayers for Biosensors" Analyst, April 1997, 122, 43R-50R
Wise, K.D.; Jackson, T.N.; Masnari, N.A.; Robinson, M.B.; Solomon, D.E.; Wuttke, G.H.; and Rensel, W.B. "Fabrication of Hemispherical Structures Using Semiconductor Technology for Use in Thermonuclear Fusion Research," Journal of Vacuum Science Technology, May/June, 1979.
Xia, Y., Whitesides, G.M., 1998. Soft Lithography. Angew. Chem. Int. Ed. 37, 550-575.
Yang, R.H., Wang, K.M., Xiao, D., Luo, K. and Yang, X.H., A renewable liquid drop sensor for di- or trinitrophenol based on fluorescence quenching of 3,3 A ,5,5 A - tetramethylbenzidine dihydrochloride, Analyst, 2000, 125, 877-882
Thus, a novel "bio-sensing" system has been shown and described. Various modifications may of course be made without departing from the sprit and scope of the invention. For example, it can be envisioned that the biosensor disclosed here can be used to sense or detect non-biological elements and compounds. The invention, therefore, should not be restricted, except by the intent of the following claims. Appendix A:
Micro Total Analysis System 2000 Manuscript Enzyme-Based Electrochemical Biosensor with DNA Array Chip
Abstract A reusable DNA sensor array for rapid biological agent detection has been fabricated on a silicon chip. (Fig.1) The DNA-based probes target the DNA/RNA sequence of the analyte instead of indirect probing using antibodies. The sensitivity is greatly enhanced by combining the hybridization event with a signal enzyme. The formation of the self-assembled monolayer sensor surface, in-situ DNA hybridization, signal measuring and the sensor regeneration can be performed within 40 minutes. Even without using the PCR, as low as 1000 Escherichia coli (E. coli) cells through 16s rRNA can be detected using this sensor array.(Fig.2) The dimension of each sensor area ranges from 25 mm2 to 160 μm2.
Keywords: Electrochemical sensor, Microelectromechanical system (MEMS), Self-assembled monolayers (SAMs), DNA hybridization
Figure imgf000028_0001
Cell number
Fig. 1 DNA sensor array chip Fig. 2 Sensitivity and specificity check
1. Introduction
The enzyme-based electrochemical biosensor is primarily motivated by the need for a highly sensitive and selective protocol capable of rapid monitoring the concentration of bacteria, virus or various biological species for field use. Such a protocol would operate remotely, and would be fully automated, compact, and robust [1]. Therefore the electrochemical transducer with minimum power consumption and smaller size is preferred over the optical system [2]. High specificity can be achieved by using DNA hybridization to reduce false positive and false negative signals. DNA electrochemical biosensors have been previously reported [3,4] using graphite or carbon electrodes. Carbon-based electrodes, however, are generally not adaptable to MEMS technology when small (<μm) dimensions are needed. In this study, Au is used as electrodes, with a protein self-assembled monolayer (SAM) to capture the E. coli rRNA. An enzyme is used as a biological amplifier in this study to gain the high sensitivity without PCR. A sensitivity and specificity check for E. coli MC4100 versus Bordetella SB54 is shown in Fig.2.
2. Detection Principle
HRP is one of the most widely used enzymes for analytical purposes and biosensors application [5]. Enzymatic amplification occurs due to a high turnover number in reactions that can be detected electrochemically. We describe here an amperometric biosensor based on horseradish peroxidase (HRP) with 3,3'5,5'-tetramethylbenzidine (TMB) as a mediator. (Fig.3) The electron transfer at the electrode surface is measured amperometrically to represent the number of the enzymes immobilized by DNA hybridization though the 16s ribosomal RNA of the target cell. Therefore, the output current is proportional to the number of the target cells in the solution.
Figure imgf000029_0001
PR : Reduced Peroxidase P0 : Oxidized Peroxidase M0 : Oxidized Mediator MR : Reduced Peroxidase
Fig. 3 Electron transport of HRP enzymatic reaction
3. Experimental
A three-electrode electrochemical cell is constructed with a micro-fabricated reaction well for the working electrode (Fig.4a). Gold (Au) is deposited as a conducting layer and Si3N4/SiO2 as an isolation layer. (Fig.4b) The surrounding Si surface is modified to be hydrophobic. The three dimensional reaction well along with the hydrophobic nature of the surrounding area allows a liquid droplet to be well contained in the working electrode. This design minimizes non-specific binding of biomolecules to other areas of the sensor chip. The Au working electrode has a monolayer of streptavidin immobilized on the surface via a thiolated-biotin SAM or through direct protein adsorption onto the gold. (Fig.4c). A sample solution containing E. coli is treated with lysis buffer, the target DNA/RNA from the E. coli cells are hybridized with both an anchoring ssDNA probe and a labelling ssDNA probe at annealing temperature (~65°C) in the presence of cell debris. (Fig.4d) The anchoring probe (conjugated to biotin) and the labelling probe (conjugated to fluorescein) recognize two distinct conservative sequences, therefore, the hybrid forms only with the specific gene segment from the target bio-agent. The oligonucleic hybrid is then immobilized through biotin-streptavidin binding onto the working electrode and unbound components are washed away. A HRP-linked anti-fluorescein antibody is then loaded onto each hybrid (Fig.4e). After addition of substrate, enzymatic reaction causes a current signal which is measured amperometrically using the three-electrode cell. (Fig.4f) The entire protocol is completed in 40 minutes.
Figure imgf000030_0001
Fig. 4 Sensor unit process flow
4. Experimental Results The SAMs on the DNA sensor were characterized by surface plasmon resonance
(SPR) and atomic force microscopy (AFM) to determine whether only a monolayer was deposited and also to determine the kinetics of protein deposition. From this data, the time scale required for in situ sensor surface formation in the integrated fluidic system can be ascertained. Two different methods were used to deposit the streptavidin SAM, one using a thiolated biotin and the other using direct protein adsoφtion. In both cases, SPR data shows that only one monolayer was deposited on the surface (Fig.5). A crystalline monolayer of streptavidin has an expected surface coverage of 2.8 ng/mm2 [6]. Our results indicate that a full monolayer was obtained using the thiolated biotin and ~80% coverage was obtained by direct protein absorption. Moreover, protein binding/adsorption occurs within ~10 seconds.
Figure imgf000031_0001
500 1000 1500 2000
Time (seconds)
Fig. 5 Surface Plasmon Resonance of Streptavidin on Bio tin- SH Au or Bare Au
Atomic Force Microscopy (AFM) was performed in the contact mode using ultralever tips with a force of 5.0nN. The Au substrate was prepared following the method of Wagner, et al. [7]. The bare Au has topographic features <lθA (Fig.όa) and contact AFM of a "full" monolayer showed a smooth surface with evidence of protein dragging (Fig.όb). The height of protein islands on a "partial" monolayer was -45 A, consistent with the dimensions of the streptavidin protein. The AFM observations confirm the SPR results that only one monolayer was deposited.
Figure imgf000031_0002
Fig. of SA ormation on u. a are u on y u coverage w t prote n dragging (c) partial coverage with protein islands.
Conclusions
The combination of MEMS technology with established DNA technology leads to a highly specific and sensitive detector for pathogenic bacteria. Biological identification using electrochemical detection with SAMs was successfully incorporated into a silicon wafer with a sensitivity that can detect less than 1 ,000 E. coli cells. References
1. Y.F. Chen, J.M Yang, J.J. Gau, CM. Ho and Y.C. Tai. "Microfluidic System for Biological Agent Detection" The 3rd International conference on the interaction of Art and Fluid Mechanics, Zurich, Switzerland , 2000. 2. D. Ivnitski, I. Abdel-Hamid, P. Atanasov, E. Wilkins, "Review: Biosensors for Detection of Pathogenic Bacteria", Biosensors & Bioelectronics 14 (1999), 599-624. 3. G. Marrazza, I. Chianella, M. Mascini, "Disposable DNA Electrochemical Biosensors for Environmental Monitoring", Analytica Chimica Acta 387 (1999), 297- 307. 4. J. Wang, et. al., "DNA Electrochemical Biosensors for Environmental Monitoring", A Review", Analytica Chimica Acta 347 (1997), 1-8. 5. T.Ruzgas, E. Csδregi, J. Emneus, L Gorton, G. Marko-Varga, "Peroxidase-modified electrodes: Fundamentals and application", Analytica Chimica Acta 330 (1996), 123- 138. 6. S.A.Darst, M. Ahlers, P.H. Meller, E.W. Kubalek, R. Blankenburg, H.O. Ribi, H. Ringsdorf, R. Kornberg, "Two-Dimensional Crystals of Streptavidin on Biotinylated Lipid Layers," Biophys J. 59 (1991) 387-396. 7. P. Wagner, M. Hegner, H-J Guntherodt, G. Semenza, "Formation and in Situ Modification of Monolayers Chemisorbed on Ultraflat Template-Stripped Gold Surfaces, " Langmuir 11 (1995) 3867-75.
ASME paper OptimiZAtion of DNA microsensor arrays for biological detection
i, Abstract
This paper describes the characterization and optimization of a reusable DNA microsensor array for rapid biological agent detection developed in previous publications.
(Fig.l) [1-3] This MEMS based DNA sensor utilizes a standard three-electrode electrochemical cell configuration with novel micro fabricated structure design to minimize non-specific binding. The sensor module is easily to be adapted to various protocols and can be used for rapid detection of macromolecules (DNA, RNA) from targets such as uropathogenic Escherichia coli (E. coli) in urine and microorganisms causing otitis media (middle ear infection). Less than 105 E. coli cells can be detected from the urine sample of a patient with urine tract infection. The sensitivity is enhanced by appropriate sensor characterization and surface modification. The total detection time including sample preparation can be reduced to 25 minutes by using a POD conjugated oligonucleotide. ii. Introduction
Conventional electrochemical detection is not quite compatible with MEMS technology and fabrication processes due to system requirements and configuration. An electrochemical cell must consist of at least two electrodes (working electrode, reference electrode) and an electrolyte. An electrochemical electrode is an interface at which the mechanism of charge transfer changes between electronic transport and ionic transport. [4] An electrolyte is a medium through which charge transfer can take place by the movement of ions. Electrochemical detection requires a second unvarying potential supplied by a reference electrode, forming a half battery. A typical reference electrode is Ag/AgCl. Currently, it is difficult to fabricate this reference electrode on a silicon substrate. Most MEMS based electrochemical sensors focus on micro fabrication of only the working electrode. This work investigates the characteristics and application of a MEMS based electrochemical sensor on silicon fabricated by standard MEMS processes. Testing of clinical urine samples with DNA hybridization on the electrochemical sensor demonstrates that MEMS based sensor on silicon can be used for biomedical detection.
iii. Detection scheme
Nucleic acid molecules (RNA/DNA) from chemically disrupted target cells are hybridized with both an anchoring ssDNA probe (conjugated to biotin) and a labeling ssDNA probe (conjugated to fluorescein) in the presence of cell debris. (Fig.2a) These two probes recognize two distinct conservative sequences, and therefore, the hybrid forms only with the specific genetic segment from the target bio-agent. The oligonucleic hybrid is then immobilized through biotin-streptavidin binding onto the working electrode and unbound components are washed away. (Fig.2b) A peroxidase (POD)-linked anti- fluorescein antibody is then loaded onto each hybrid. (Fig.2c) After addition of substrate with a mediator (tetramethylbenzidine, TMB), enzymatic reaction causes a current signal which is measured amperometrically from the REDOX reaction shown in Fig.3. The entire protocol can be completed in 40 minutes and the sensor reactivated after a cleaning process.
iv. Sensor characterization Most biosensor chips utilize off-chip optical detection. Our work is directed at on- chip electrochemical detection with sensor optimization and surface modification for signal-noise-ratio improvement. MEMS technology makes possible the development of miniaturized electrochemical cells since conductive metallic electrodes in small dimensions can be accurately deposited and patterned on a silicon substrate. From our studies, Au was a suitable candidate for all three electrodes (working, auxiliary, and reference), and characterization of the Au/Au/Au electrode cell using a well-known one- electron system such as ferrocene confirmed that classic reversible one-electron transfer was obtained.
v. Cyclic voltammetry Cyclic voltammetry for reversible one-electron transfer of ferrocene is characterized by a peak separation of ~57 mV between the anodic and cathodic peaks, the same peak maximum, and a linear relationship between peak current vs. [scan rate] [4]. As seen in Figure 4, voltammograms of ferrocene using MEMS based electrochemical sensor at different scan rates show the expected behavior and a plot of the peak current at
1 /9 • peak maximum vs. [scan rate] is linear. Further characterization of the electrochemical cell using peroxidase confirmed that enzymatic amperometry is also feasible. Cyclic voltammetry conducted on the mediator shows a two-electron redox transfer, as expected for TMB (Fig 5), and addition of the peroxidase enzyme results in an increase in the reduction current that can be clearly measured at -0.1V bias potential.
vi. Reduction of non-specific bonding
The sensor surface has a protein self-assembled monolayer to capture the target RNA/DNA onto the working electrode, and the SAMs have been characterized by atomic force microscopy (AFM) and surface plasmon resonance (SPR). [2] The signal-to-noise can be improved significantly by appropriate surface modifications to reduce non-specific binding. The first approach is to introduce a surface blocking protein to eliminate potential bonding sites for non-specific binding. (Fig 6) The second approach is adding a hydrophobic modification of the periphery region with silanation treatment or thin-film coatings of Teflon® or polyimide. With the hydrophilic nature of protein-covered Au working electrode, reagents will be confined to the working electrode only, minimizing deposition of enzyme to the periphery and other two electrodes. (Fig 7,8) In both cases, noise due to non-specific binding was reduced. Typically, the second approach (hydrophobic modification) is used in MEMS fabrication and the first one is incorporated into probe reagent.
vii. Protocol simplification Sample preparation is the most time consuming step for most biosensors. Any additional protocol steps will require more fluidic devices and a longer detection time. A simplified protocol is shown in Fig.9 whereby the step of the enzyme loading with POD conjugation onto detector DNA probe is eliminated. The hybridization condition was controlled to retain the enzymatic activity of POD after heating to 65°C. The protocol can then be shortened fro m 40 to 25 minutes while reducing the required number of reagents by one.
viii. Result
Previous work has demonstrated the sensitivity of micro DNA sensor to be -10 E. coli cells. Recent data show it has the capability to detect less than 10 E. coli cells from cell culture at stationary stage through ribosomal RNA content. The sensor was then subjected to clinical urine sample from Division of Infectious Disease at UCLA for testing. As shown in Figure 10, the minimum detectable cell number of E. coli in urine sample is 105 from our preliminary result without isolation. Sensitivity with urine sample can be improved with further optimization. Another type of bacteria, Bordetella, was used as a negative control and all E. coli samples, including dilutions, have higher signal than the negative control.
Micro DNA sensor can be used for electrochemical detection without utilizing a conventional reference electrode because the bias potential is reasonable low (-0.1V) and the detection is short enough (20 seconds) to avoid the accumulation of charge at auxiliary electrode. This was confirmed with voltammograms of ferrocene and POD solution.
We have demonstrated that micro DNA sensor can be used for electrochemical detection of pathogens such as E. coli. The Au/Au/Au three electrode cells patterned on silicon by MEMS technology were successfully used in amperometric measurement of enzymatic reactions. These sensors show promise that they can be incoφorated into micro total analysis system (μ-TAS) or "Lab on a Chip".
ix. Conclusion
A micro DNA sensor was fabricated that can detect enzymatic reaction amperometrically and was characterized by using cyclic voltammetry with ferrocene and POD to demonstrate the capability of conducting electrochemical detection without a conventional Ag/AgCl electrode. Noise reduction was accomplished by surface modification and introducing blocking protein. The structural design of a well in the working electrode and surface treatment on silicon substrate enable reagent confinement over the working electrode. The reagent-electrode contact can be controlled to reduce non- specific binding, not possible with a conventional beaker setup. Urine sample testing shows this DNA sensor is suitable for clinical diagnosis with a short detection time and smaller system size. References Chen, Y F., Yang, J.M., Gau, J.J., Ho, CM., Tai, Y.C, 2000. "Microfluidic System for Biological Agent Detection" Proceedings of the 3rd International conference on the interaction of art and fluid mechanics, Zurich, Switzerland, 2000. Gau, J.J., Lan, E. H., Dunn, B., Ho, CM., 2000. "Enzyme-based electrochemical biosensor with DNA array chip" Proceedings of the fourth International Symposium on Micro Total Analysis Systems (μTAS), Enschede, The Netherlands. Gau, J.J., Lan, E. H., Dunn, B., Ho, CM., 2000. "A MEMS Based Amperometric Detector for E. Coli Bacteria - Using Self-Assembled Monolayers" Proceedings of the sixth World Congress on Biosensors, San Diego, USA Eggins, B., 1996 Biosensors, John Wiley and Sons, New York. Hall, E. A. H., Biosensors, Prentice Hall, Eaglewood Cliffs, NJ, pp. 97-139, 1991
Figure imgf000036_0001
Fig.l DNA microsensor array chip
(a) 4 sensors with surface feature to increase surface-volume -ratio
(b) 14 sensors in 1cm2
Figure imgf000037_0001
Fig.2 Sensor and protocol process flow (a) hybridization (b) immobilization (c) POD loading
Electrode
Figure imgf000037_0002
PR . Reduced Peroxidase , P0 : Oxidized Peroxidase .-,. „ „, Mo' Oxidized Mediator. , MR . Reduced Mediator
Fιg.3 Electron transfer of enzymatic reaction of horseradish peroxidase
Figure imgf000038_0001
Fitr.4 Sensor characterization bv cvclic voltammetrv
Figure imgf000038_0002
Potential /V Fig.5 Sensor characterization with HEP enzvmatic
Figure imgf000038_0003
Amperometiy Detection Time (10 sec)
^o blocking protein ■with Bovine Serum Albumin (BSA) D with KPL Detector Bbck© Fjgδ Noise reduction by adding blocking proteins to eliminate non-specific binding
Figure imgf000039_0001
Fig.7 Comparison of electrode-reagent contact between (a) conventional and (b) micro DNA sensor
Figure imgf000039_0002
Fig.8 Image of electrolyte confinement over electrodes of micro DNA
Figure imgf000040_0001
Fig.9 Simplified protocol with POD conjugated labeling probe (a) hybridization (b) immobilization
Figure imgf000040_0002
E. coli cell number in urine sample
Fig.10 Sensitivity check of micro DNA sensor with urine sample A MEMS BASED AMPEROMETRIC DETECTOR FOR E. COLI BACTERIA USING SELF-ASSEMBLED MONOLAYERS
Abstract
We developed a system for amperometric detection of Escherichia coli (E. coli) based on the integration of microelectromechanical systems (MEMS), self-assembled monolayers (SAMS), DNA hybridization, and enzyme amplification. Using MEMS technology, a detector array was fabricated which has multiple electrodes deposited on a Si wafer and was fully reusable. Using SAMs, a monolayer of the protein streptavidin was immobilized on the working electrode (Au) surface to capture rRNA from E. coli. Three different approaches can be used to immobilize streptavidin onto Au, direct adsorption of the protein on bare Au, binding the protein to a biotinylated thiol SAM on Au, and binding the protein to a biotinylated disulfide monolayer on Au. The biotinylated thiol approach yielded the best results. High specificity for E. coli was achieved using ssDNA-rRNA hybridization and high sensitivity was achieved using enzymatic amplification with peroxidase as the enzyme. The analysis protocol can be conducted with solution volumes on the order of a few microliters and completed in 40 minutes. The detection system was capable of detecting 1000 E. coli cells without polymerase chain reaction with high specificity for E. coli vs. the bacteria Bordetella bronchiseptica.
Key words: Microelectromechanical systems (MEMS), self-assembled monolayers (SAMs), amperometric detection, Escherichia coli bacteria, DNA hybridization
1. Introduction
A variety of biosensors, both optical and electrochemical, have been developed for the detection of pathogenic bacteria (Ivnitski, et al., 1999, 2000). While conventional methods for detecting bacteria usually involve a morphological evaluation of the organisms as well as testing their ability to grow, such methods are very time consuming and are not feasible under field conditions. The need for rapid detection as well as portability has led to the development of systems that couple pathogen recognition with signal transduction. Both optical and electrochemical detection of bacteria have been reported, although electrochemical methods have an advantage in that they are more amenable to miniaturization (Ivnitski, et. al., 1999, T. Wang, et. al., 2000). Requirements for an ideal detector include high specificity and high sensitivity using a protocol that can be completed in a relatively short time. Moreover, systems that can be miniaturized and automated offer a significant advantage over current technology, especially if detection is needed in the field.
Microelectromechanical systems (MEMS) technology provides transducers to perform sensing and actuation in various engineering applications. The significance of MEMS technology is that it makes possible mechanical parts of micron size that can be integrated with electronics and batch fabricated in large quantities. MEMS devices are fabricated through the process of micromachining, a batch production process employing lithography. Micromachining relies heavily on the use of lithographic methods to create 3 -dimensional structures using pre-designed resist patterns (or masks) and then selectively etching the undesirable parts away (Ho and Tai, 1996, 1998). MEMS is an enabling technology for making miniaturized devices, and in this work, we integrate MEMS technology with biosensing methods to detect E. coli bacteria.
One of the most effective means of achieving high specificity is to detect the bacteria's genetic material (e.g. rRNA, mRNA, denatured DNA). By choosing a single- stranded DNA (ssDNA) probe whose sequence is complementary only to the target bacteria's rRNA or ssDNA, monitoring the hybridization event allows selective sensing of target cells. To maximize sensitivity, coupling the hybridization event with an enzymatic reaction leads to signal amplification, as each substrate-to-product turnover contributes to the overall signal. Bioassays to detect DNA hybridization that are amplified by enzymatic reaction can still be completed within a reasonably short time. Finally, the miniaturization and portability inherent in electrochemical probes make them excellent candidates for incorporation in MEMS devices.
We developed a prototype amperometric detector for Escherichia coli (E. coli) based on combining several well-established technologies into one detection system (Chen, et. al., 2000, Gau, et. al., 2000). The technologies of MEMS, self-assembled monolayers (SAMs), DNA hybridization, and enzyme amplification all contribute to the design of a miniaturized, specific, and sensitive E. coli detector. DNA electrochemical probes have been reported previously (Wang, et al., 1997, Marrazza, et al., 1999), with graphite or carbon electrodes typically used. Commercial units for amperometric detection of DNA from E. coli using screen-printed carbon electrodes on disposable test strips are also available (Andcare Inc., Durham, NC). Screen-printing has the advantage of low cost, but achieving high dimensional precision is not easy . Using lithography, thin films of a wide range of materials, including metals (Au, Ag) and carbon, can be accurately patterned in μm size dimensions. Moreover, SAMs is an elegant method of selectively immobilizing molecules on MEMS surfaces. The bonding of SAMs to Au, Ag and other metals has been well studied (Revell, et al., 1998, Motesharei, et al, 1998, Xia, et al., 1998, Lahiri, et al., 1999), and proteins and other biomolecules can be easily immobilized onto surfaces such as Au using SAMs (Ostuni, et al., 1999, Kane, et al., 1999, Spinke, et al., 1993, Haussling, et al., 1991). Moreover, amperometric methods using SAMs on electrodes have demonstrated the ability to detect target analytes successfully (Sun, et al., 1998, Hou, et al., 1998, Murthy, et al., 1998). In our E. coli detection system, we take advantage of the benefits inherent in each technology. Using DNA hybridization and enzyme amplification, we achieve the required specificity and sensitivity. Using MEMS and SAMs, we fabricate a miniaturized system that can lead to a portable instrument. Finally, we demonstrate that the present detection system is applicable to a broad range of pathogenic bacteria. For example, the detection module and assay protocol can be adapted to detect uropathogenic E. coli and identification of microorganisms causing otitis media (middle ear infection).
2. Experimental
2.1. MEMS detector array
A layer of silicon dioxide (SiO2, lOOOA) was deposited on a bare Si wafer (prime grade, p-type <100>, thickness 500-550μm) and served as a pad layer underneath the silicon nitride (Si3N , 1000 A) to release stress and improve adhesion. MEMS arrays were fabricated with working electrodes of 3.6mm x 3.6 mm etched to form wells of 350μm depth. The nitride wafer was patterned and bulk etched using KOH along [111] and [100] crystal planes, and depth of the well was controlled by KOH etching time and temperature. The lOOμm wide auxiliary and reference electrodes are separated from their corresponding working electrode by 200μm. Figure 1 shows a schematic of the pattern used in generating the MEMS array. The nitride and oxide were removed by HF etching to release internal stress, and another oxide layer (5000A) was deposited for electrical isolation. Electrodes were patterned by PR5214 photo resist reverse imaging and lift-off process with e-beam deposition of Au(200θA)/Cr(20θA). Finally the wafer was bathed in hexamethyldisilazane (HMDS) vapor for three minutes after ten minutes of a 150°C hot bake to generate a hydrophobic surface on the surrounding Si areas. The hydrophobic nature of the surrounding area along with the 3 -dimensional nature of the working electrode allows containment of a liquid droplet in the working electrode. This design effectively minimized non-specific binding of biomolecules to other areas of the MEMS array.
2.2. Deposition of streptavidin SAMs on Au
Three different methods were used to deposit streptavidin monolayers on Au: 1) directly adsorbing streptavidin on bare Au, 2) depositing a SAM of a biotinylated thiol, biotin-DAD-C12-SH, and subsequently binding streptavidin, 3) depositing a SAM of a biotinylated disulfide, biotin-HPDP, and subsequently binding streptavidin. In all cases, the Au surfaces were cleaned with concentrated "Piranha" solution (70 vol% H2SO , 30 vol% H2O2) and thoroughly rinsed with deionized water. For depositing streptavidin on bare Au, a solution of 1.0 mg/ml streptavidin (Sigma Chemical Co., S0677) in 0.02M Na phosphate buffer, 0.15M NaCl, pH 7.2, was placed on the surface, allowed to stand for 10 minutes, and rinsed with deionized water. For depositing a SAM of biotin-DAD-C12-SH (12-mercapto(8-biotinamide-3,6-dioxaoctyl)dodecanamide, Roche GmBH, Germany), the procedure of Spinke, et al. (1993) was used wherein samples were incubated for -18 hours in a 50μM solution of biotin-DAD-C12-SH in ethanol with 4.5x10" 11-mercapto-l- undecanol (Aldrich Chemical Co., 44,752-8) and rinsed with ethanol and water. The biotin-coated Au surfaces were then exposed to a 1.0 mg/ml streptavidin solution for —10 minutes and rinsed again with water. For depositing a SAM of biotin-HPDP, (N-[6- (biotinamido)hexyl]-3'-(2'-pyridyldithio)propionamide, Pierce Inc., 21341) samples were incubated for -18 hours in a 50μM biotin-HPDP solution in ethanol (with or without 4.5xl0"4M mercaptopropanol) and rinsed with ethanol and water. The surfaces were then exposed to a 1.0 mg/ml streptavidin solution for -10 minutes and rinsed again with water.
2.3 Assay protocol for amperometric detection of E. coli
The assay protocol was conducted as follows: 1) 50 μl of lysis reagent (Andcare Inc., 4002-11) was added to a 250 μl sample of bacteria in culture media and incubated for 5 min. at room temperature, 2) 100 μl of probe solution (Andcare Inc., 4002-13) was then added and the mixture was incubated for 10 min. at 65°C, 3) 5 μl of the lysed E. coli/probe solution mixture was placed on the streptavidin coated working electrode of the MEMS detector array and incubated for 10 min. at room temperature, 4) the MEMS detector array was washed with biotin wash solution (Kirkegaard and Perry Laboratories, 50-63-06), 5) 5 μl of Anti-Fl-POD (Anti-fluorescein peroxidase, 150U, Roche Inc., 1 426 346), diluted to 0.75U/ml or 0.15 U/ml with diluant (Andcare Inc., 4002-14) was placed on the working electrode and incubated for 10 minutes at room temperature, 6) the MEMS array chip was washed again with wash solution, 7) 10 μl of K-blue substrate (Neogen Corp., 300176) was placed on the detector array in such a way that all three electrodes (working, auxiliary, reference) were covered by the substrate solution, and 8) electrochemical measurements were immediately taken. The entire protocol was completed within 40 minutes. Amperometric current vs. time was measured using a CH Instruments 660A electrochemical workstation with picoamp booster and faraday cage. Samples on the MEMS detector array were measured sequentially. The voltage was fixed at -0.1V (vs. reference), and the cathodic current at 20 seconds was taken as the amperometric signal. At 20 seconds, current values reached steady-state. Cell concentration (cell number) was determined using serial dilutions and culture plate counting.
2.4 Characterization of Self- Assembled Monolayers
The performance of the detector depends heavily on the properties of the immobilized streptavidin monolayer. We performed surface plasmon resonance (SPR, Biacore X system, Biacore, Inc.) and atomic force microscopy (AFM) to characterize the monolayers. For SPR studies of streptavidin binding to bare Au, bare Au chips (Jl sensor chips) were used. For studies of streptavidin binding to biotin-DAD-C-12-SH/Au or biotin-HPDP/Au, the biotin SAM was deposited on the bare Au chips as previously described before the SPR experiments. For best results, new chips were cleaned with diluted H SO /H2O2 solution for -2 min. before performing SPR with bare Au or depositing the biotin SAM. In all cases, 1.0 mg/ml streptavidin in 0.02M Na phosphate, 0.15 M NaCl, pH 7.2 buffer was used. In adsoφtion experiments with streptavidin on bare Au, flow rates ranged from 1 to 5 μl/min. In the experiments with streptavidin and biotin-DAD-C12-SH/Au, flow rates ranged from 10 to 25 μl/min. In experiments with streptavidin and biotin-HPDP/Au, flow rates ranged from 5 to 10 μl/min. In the desorption experiments, 25 μl of the following solutions were flowed sequentially through the channels at 25 μl/min (total exposure time of 1 min.): 1.0M KC1, 8M urea, 0.5% SDS, 0.1M HC1, 0.1M NaOH, and 40 vol% formamide. AFM (AutoProbe CP, Thermomicroscopes, Inc.) was performed in the contact mode using ultralever tips with a force of 5.0nN. To ensure a flat Au surface, the method of Wagner, et al. (1995) was used wherein Au was first deposited via e-beam evaporation on mica and then transferred to Si. In our case, the mica was cleaved to cleanly remove it from the Au without the use of solvent.
3 Results
3.1 Microelectromechanical system (MEMS)
Figure 2 shows a photograph of the MEMS detector array. Sixteen working electrodes with their corresponding auxiliary and reference electrodes were patterned in a 2.8 cm x 2.8 cm area. The detector array was fully reusable as the surface can be cleaned using H2SO /H2O2 solutions. We have reused the same MEMS detector array multiple times by appropriately cleaning the surface and redepositing the SAMs on the working electrode. 3.2 E. coli Detection Using DNA Hybridization
In our MEMS system, E. coli detection is based on DNA hybridization followed by enzymatic reaction. A schematic illustrating the electrode surface is shown in Figure 3. A streptavidin monolayer is immobilized on the Au working electrode surface to capture the rRNA from E. coli. Two ssDNA segments are used in this system. The capture ssDNA, which is conjugated to biotin for streptavidin binding, hybridizes to one end of the E. coli rRNA. The detector ssDNA, which is conjugated to fluorescein for binding to anti- fluorescein linked to the enzyme peroxidase (POD), hybridizes to the other end of the E. coli rRNA. The capture and detector ssDNA recognize two distinct conservative sequences, and therefore, the hybrid forms only with the specific gene segment from E. coli. The oligonucleic hybrid is immobilized through biotin-streptavidin binding onto the Au working electrode and unbound components are washed away. Streptavidin binds biotin with unusually high affinity (Kd -10"15M) (Weber, et. al., 1989). After loading the POD onto the hybrid (through Anti-Fl-fluorescein binding), substrate is added and enzymatic reaction is detected amperometrically. The substrate solution contains both the substrate H2O2 and a mediator, 3,3',5,5'tetramethylbenzidine (TMB). The enzyme and electrode reactions are depicted in Figure 4.
3.3 Streptavidin Self Assembled Monolayers for rRNA Capture
We used three different approaches to immobilize a streptavidin monolayer on the electrode surface, as shown in Figure 5. In the first approach, a streptavidin monolayer was deposited on bare Au via protein adsorption. In the second approach, a SAM of biotin was deposited on the Au using a biotinylated thiol and streptavidin was subsequently bound to the biotin. In the third approach, a SAM of biotin was deposited on the Au using a biotinylated disulfide and streptavidin was subsequently bound to the biotin.
3.4 Characterization of Streptavidin Monolayers
The streptavidin monolayers were characterized using both surface plasmon resonance (SPR) and atomic force microscopy (AFM). SPR has been demonstrated to be a viable technique for monitoring interactions of molecules with metallic (Au, Ag) thin films at the solution-metal interface. This technique can be used to estimate the thickness of a deposited layer as well as to measure the kinetics of association and dissociation (Haussling, et al., 1991, Spinke, et al., 1993, Sigal, et al., 1996, Rao, et al., 1999, Jung, et al., 1999). We performed SPR to monitor deposition of streptavidin on Au using all three approaches. SPR results are shown in Figure 6. Based on calibrations by the manufacturer (Biacore), 1000 resonance units (RU) in the SPR signal is equivalent to a change of -1 ng/mm2 in surface protein concentration. Streptavidin has dimensions of - 55 x 45 x 50 A (Darst, et al., 1991). A full monolayer of streptavidin has an expected density of -2.8 ng/mm2, calculated based on a 2-dimensional crystalline monolayer (Jung, et al., 1999, Darst, et al., 1991). From Figure 6, essentially a complete monolayer of streptavidin was deposited on the biotinylated thiol SAM/Au (-3000 RU), -80% "coverage" was obtained with streptavidin deposited on bare Au (-2400 RU), and -52% "coverage" was obtained with streptavidin deposited on the biotinylated disulfide SAM/Au (-1550 RU). For the biotinylated disulfide, the presence of mercaptopropanol in the solution had a negligible effect on surface coverage as SPR signal increased only -10% (-1700 RU, data not shown). In all cases, additional protein deposition upon a second injection of protein solution was minimal. Moreover, flow rates had negligible effects on the rate or amount of protein deposited. The SPR results indicate that in all three approaches, only a monolayer of streptavidin, and not multilayers, was deposited on the Au. Finally, these results establish that most of the streptavidin-biotin binding and streptavidin adsoφtion on bare Au occurs within seconds and can be completed on the order of minutes.
We performed experiments to determine whether streptavidin can be desorbed, i.e. whether streptavidin can be dissociated from the bare Au or from binding to biotin to regenerate the surface. Table I lists the loss in SPR signal (RU) after treatment with various reagents that are known to dissociate protein-ligand binding and/or denature proteins. As seen in Table I, only 8M urea, 0.5% SDS, and 0.1M NaOH were somewhat effective in desorbing streptavidin from the surface. 1.0M KC1, 0.1M HC1, and 40% formamide were not effective. Streptavidin could not be completely desorbed by any of the reagents, and some protein remained after subjecting the surface to all reagents. These results show that streptavidin had rather good binding to both the biotin SAM as well as to bare Au and that the streptavidin monolayers were relatively stable.
The use of AFM enables us to further characterize the surface by imaging streptavidin directly adsorbed on bare Au. Bare Au had topographic features <10 A, whereas protein islands on a "partial" monolayer was -45 A, consistent with the dimensions of streptavidin (Figure 7). The AFM results confirm the SPR findings that only one monolayer was deposited on the Au. AFM in the contact mode for a "full" protein monolayer showed a featureless surface with evidence of protein dragging (data not shown), and therefore, did not provide any additional information as to surface coverage. As previously reported, monolayer deposition is also obtained when using biotinylated SAMs and subsequently binding streptavidin (Spinke, et. AL, 1993, Jung, et. AL, 1999). 3.5 Electrochemical Measurements with MEMS Detector Array
In our MEMS detector array, we used a three-electrode system with Au for all three electrodes, i.e. working, auxiliary, and reference electrodes. Typically, Ag/AgCl or saturated calomel electrode (SCE) is used as the reference electrode so that reversible oxidation/reduction at fixed potential occurs at the reference electrode. In our MEMS detector array, however, we used Au as the reference electrode to simplify fabrication and to permit a fully reusable array. Maintaining a constant potential is made possible by the use of a 3-electrode system (vs. a 2-electrode system). In our particular application where the reduction of TMB was monitored, Au can be successfully used as the reference electrode because a low voltage difference (-0.1V) was maintained for short periods of time (<1 min).
We characterized the Au/Au/Au electrode system for electrochemical detection by two separate experiments. In the first experiment, a ferrocene film was placed on the electrodes and cyclic voltammetry was conducted to monitor the redox reactions. Cyclic voltammetry for a classic reversible one electron transfer is characterized by a peak separation of -57 mV between the anodic and cathodic peaks, the same peak currents at
11 peak maximum, and a linear relationship between peak current vs. [scan rate] (Hall, 1991). Figure 8 shows the voltammograms obtained with ferrocene at different scan rates. As seen in Fig. 8, classical redox behavior was observed and a plot of the peak current vs. [scan rate]1 2 is linear (Figure 9).
In a second experiment with the Au/Au/Au electrode system, cyclic voltammetry was conducted on the substrate solution only (H2O2 + TMB) and then again on the same solution with POD enzyme. Cycling between -0.2V to +0.50V at a scan rate of 10m V/s, the substrate solution showed the two electron redox behavior of TMB (Figure 10). The addition of POD to the substrate solution resulted in an increase in the reduction current. A constant potential of-O.lOV (vs. reference) was then selected for measurement of POD enzymatic activity. At this potential, the current background was near zero and no substrate oxidation occurred. This potential was optimum for enzymatic activity determination in which a small amount of product (oxidized TMB) was to be measured in the presence of high concentrations of substrate.
3.6 Detection of E. coli
For the amperometric detection of E. coli rRNA, we first compared the performance of the three different streptavidin monolayer surfaces. Streptavidin was immobilized on the Au using the three different approaches previously described, and the assay protocol was conducted for the bacteria E. coli and Bordetella bronchiseptica
(Bordetella). Since the ssDNA probes are specific for E. coli, the Bordetella bacteria served as the negative control sample. The puφose of this experiment was to compare the efficacy of the immobilized streptavidin to capture the biotin-rRNA-POD hybrid. Two concentrations of E. coli were used, with one sample having ten times the concentration of the other. Moreover, the signal from the Bordetella indicates the level of non-specific binding or the achievable "baseline". Results from this experiment are shown in Figure 11. Since the same bacterial solutions (E. coli or Bordetella) were used, a direct comparison can be made of the different surfaces. As seen in Fig. 11, streptavidin immobilized via the biotinylated thiol to Au was the best condition for E. coli detection. Using the biotin-thiol SAM, we obtained good signals for the E. coli while achieving a low baseline signal from the Bordetella. For streptavidin immobilized via the biotin- disulfide to Au, current signals for the E. coli (both concentrations) were significantly lower, while the baseline was the same as that for the biotin-thiol/streptavidin. In the case of streptavidin directly adsorbed to Au, the signal from the Bordetella was much higher, indicating a higher level of non-specific binding of POD to the surface. After ascertaining Au/biotin-SH/streptavidin to be the optimal streptavidin surface, we repeated the protocol using E. coli and Bordetella to determine the sensitivity of our system. We immobilized streptavidin via the biotin-SH SAM and performed the assay on a series of E. coli dilutions along with Bordetella as the negative control. Results are shown in Figure 12. The data indicate that as few as 1000 E. coli cells can be detected using our MEMS system. As expected, current signal increased as a function of increasing number of E. coli cells in the sample solution. Moreover, by lowering the POD concentration used in the assay protocol (from 0.75 U/ml to 0.15 U/ml), we achieved better discrimination in signals at lower E. coli cell numbers (Figure 13). As seen in Fig. 13, the current signal for 1000 E. coli cells was more than twice that for 2.5xl05 Bordetella cells. The results using our MEMS system confirm that E. coli bacteria was successfully detected using amperometry and SAMs to capture the bacteria rRNA.
4. Discussion
Our results show that combining MEMS technology with SAMs, DNA hybridization, and enzymatic amperometry leads to a highly specific and sensitive electrochemical detector for bacteria such as E. coli. The contribution from each component is critical to the overall success of the system. MEMS technology enables an array of multiple three-electrode "cells" to be deposited on a Si wafer, and the MEMS detector array we have described is fully reusable as the SAMs can be removed and the Au surfaces regenerated with appropriate cleaning. Moreover, with micromachined channels, valves, pumps and integrated electronics, one can fully automate the sample preparation and assay protocol. SAMs provide an effective means of functionalizing the Au working electrode to immobilize capture biomolecules, for example, streptavidin. DNA hybridization permits high specificity for pathogenic bacteria as the sequence of the ssDNA probes can be carefully selected to complement only the target. Coupling the hybridization event with an enzymatic reaction provides signal amplification and enhances the sensitivity. Finally, by using electrochemical transduction, a miniaturized portable system with minimum power consumption can be developed.
Rapid detection and a portable instrument is desirable for pathogen sensing. We have demonstrated that detection can be achieved within -40 minutes using this system. Due to the small dimensions and small sample volumes, it is possible to further reduce the assay time by reducing the incubation times (10 minutes) currently used for DNA hybridization and enzyme binding. A distinct advantage of MEMS is the ability to use very small volumes (a few μl) and electrode surface areas (currently 0.13 cm2 for the working electrode, <0.02 cm for the auxiliary and reference electrodes). Our results show that as few as -103 cells can be detected using this system without polymerase chain reaction (PCR). Due to the small volumes and working electrode surface area in the MEMS system, reporting the detection limit in terms of absolute cell numbers is more appropriate than reporting the detection limit in terms of cell concentration (cells/ml). Detection limits on the order of 102 to 103 cells/ml have been reported, however, sample volumes of - 1.0 ml with working electrode surface areas -1cm were typically used (Abdel-Hamid, et. al., 1998, 1999). In amperometric enzyme immunofiltration assays,, the signal was more than an order of magnitude less when using a 0.1 ml sample than when using a 1.0 ml sample (Abdel-Hamid, 1999).
Results from amperometric experiments to detect E. coli rRNA have shown that the streptavidin monolayer immobilized via the biotinylated thiol SAM approach yielded the best results. This finding is not suφrising as SPR data indicated the highest streptavidin surface density or "coverage" when using the biotinylated thiol. It is likely that a well-ordered self-assembled monolayer is formed only with the biotinylated thiol, leading to highest streptavidin surface density. In the case of the biotinylated disulfide, although attachment of the biotin to the Au surface occurs via the Au-S bond, the additional organic group probably hinders the formation of a densely packed monolayer. Streptavidin immobilized via direct adsoφtion to Au resulted in significantly higher nonspecific binding of the POD enzyme to the working electrode. Protein adsoφtion to Au has been well known as colloidal Au particles attached to various proteins (e.g. streptavidin, immuno globulins) are commercially available (Nanoprobes, Inc., Yaphank, NY). Streptavidin does not contain cysteine or methionine residues (Weber, et. al., 1989) and therefore, does not attach to Au via an Au-S bond. Protein adsoφtion to Au can occur via interaction of carboxylate groups with Au (Ooka, et. al., 1999) and is the likely mechanism for streptavidin-Au attachment. Although a streptavidin monolayer can be attached to Au via direct adsoφtion, whether self-assembly or molecular ordering occurs is questionable. SPR desoφtion experiments showed the streptavidin-Au attachment to be as robust as streptavidin-biotin binding, i.e. the amount of streptavidin removed due to urea, SDS, and NaOH were similar for streptavidin directly adsorbed on Au as compared to streptavidin attached to biotin. When conducting the assay protocol for E. coli, however, sample solutions contained oligonucleotides as well as cell debris from the lysed E. coli. Our data suggests that the presence of other proteins and biomolecules accelerated the desoφtion of streptavidin from Au, leading to increased non-specific binding of the enzyme POD to the surface.
5. Conclusions
E. coli bacteria were successfully detected by incoφorating MEMS with SAMs, DNA hybridization, and enzyme amplification. We demonstrated a MEMS-based detection system that is specific for E. coli and capable of detecting 1000 cells without PCR. The process time can be 40 minutes or less. Moreover, the assay can be conducted with solution volumes on the order of a few microliters. The integration of SAMs, DNA hybridization, and enzyme amplification methodologies with MEMS technology makes possible a new generation of devices for pathogenic detection.
References
Abdel-Hamid, I. Ivnitski, D., Atanasov, P., Wilkins, E., 1998. Fast Amperometric Assay for E. coli 0157:H7 using partially immersed immunoelectrodes. Electroanalysis 10 (11), 758-763.
Abdel-Hamid, I., Ivnitski, D., Atanasov, P., Wilkins, E., 1999. Flow-through immunofiltration assay system for rapid detection of E. coli 0157:H7. Biosens. Bioelect. 14, 309-316.
Chen, Y.F., Yang, J.M., Gau, J.J., Ho, CM., Tai, Y.C, 2000. Microfluidic System for Biological Agent Detection. Proceedings of the 3rd International conference on the interaction of art and fluid mechanics, Zurich, Switzerland. Darst, S.A., Ahlers, M., Meller, P.H., Kubalek, E.W., Blankenburg, R., Rib, H.O.,
Ringsdorf, H., Kornberg, R.D., 1991. Two-dimensional crystals of streptavidin on biotinylated lipid layers and their interactions with biotinylated macromolecules. Biophys. J. 59, 387-396. Gau, J.J., Lan, E. H., Dunn, B., Ho, CM., 2000. Enzyme-based electrochemical biosensor with DNA array chip. Proceedings of the fourth International Symposium on Micro Total Analysis Systems (μTAS), Enschede, The Netherlands.
Hall, E.A.H., 1991. Biosensors. Prentice Hall, Englewood Cliffs, New Jersey. Haussling, L., Ringsdorf, H., Schmitt, F.J., Knoll, W., 1991. Biotin-functionalized self-assembled monolayers on gold: surface plasmon optical studies of specific recognition reactions. Langmuir 7 (9), 1837-1840.
Ho, CM., Tai, Y.C, 1996. Review: MEMS and its applications for flow control. J. Fluids Eng. 118, 437-447. Ho, CM., Tai, Y.C, 1998. Micro-electro-mechanical systems (MEMS) and fluid flows. Ann. Rev. Fluid Mech. 30, 579-612.
Hou, S.F., Yang, K.S., Fang, H.Q., Chen, H.Y., 1998. Amperometric glucose enzyme electrode by immobilizing glucose oxidase in multilayers on self-assembled monolayers surface. Talanta 47, 561-567. Ivnitski, D., Abdel-Hamid, I., Atanasov, P., Wilkins, E., 1999. Biosensors for detection of pathogenic bacteria. Biosens. Bioelect. 14, 599-624.
Ivnitski, D., Abdel-Hamid, I., Atanasov, P., Wilkins, E., Strieker, S., 2000. Application of Electrochemical Biosensors for Detection of Food Pathogenic Bacteria. Electroanalysis 12 (5), 317-325. Jung, L.S., Nelson, K.E., Campbell, C.T., Stayton, P.S., Yee, S.S., Perez-Luna, V.,
Lopez, G.P., 1999. Surface plasmon resonance measurement of binding and dissociation of wild-type and mutant streptavidin on mixed biotin-containing alkylthiolate monolayers. Sensors Actuators B 54, 137-144.
Kane, R.S., Takayama, S., Ostuni, E., Ingber, D.E., Whitesides, G.M., 1999. Patterning proteins and cells using soft lithography. Biomaterials 20, 2363-2376.
Lahiri, J., Ostuni, E., Whitesides, G.M., 1999. Patterning Ligands on Reactive SAMs by Microcontact Printing. Langmuir 15 (6), 2055-2060.
Marrazza, G., Chianella, I., Mascini, M., 1999. Disposable DNA electrochemical biosensors for environmental monitoring. Anal. Chim. Acta 387, 297-307. Motesharei, K., Myles, D.C., 1998. Molecular recognition on functionalized self- assembled monolayers of alkanethiols on gold. J. Am. Chem. Soc. 120 (29), 7328-7336.
Murthy, A.S.N, Sharma, J., 1998. Glucose oxidase bound to self-assembled monolayers of bis(4-pyridyl) disulfide at a gold electrode: Amperometric determination of glucose. Anal. Chim. Acta 363, 215-220. Ooka, A. A., Kuhar, K.A., Cho, N., Garrell, G.L., 1999. Surface Interactions of a
Homologous Series of a, w- Amino Acids on Colloidal Silver and Gold. Biospectroscopy 5, 9-17. Ostuni, E., Yan, L., Whitesides, G.M., 1999. The interaction of proteins and cells with self-assembled monolayers of alkanethiolates on gold and silver. Colloids Surfaces B 15, 3-30.
Rao, J., Yan, L., Xu, B., and Whitesides, G.M., 1999. Using surface plasmon resonance to study the binding of vancomycin and its dimer to self-assembled monolayers presenting D-Ala-D-Ala. J. Am. Chem. Soc. 121 (11), 2029-2030.
Revell, D.J., Knight, J.R., Blyth, D.J., Haines, A.H., Russell, D.A., 1998. Self- assembled carbohydrate monolayers: formation and surface selective molecular recognition. Langmuir 14 (16) 4517-4524. Sigal, G.B., Bamdad, C, Barberis, A., Strominger, J., and Whitesides, G.M., 1996.
A self-assembled monolayer for the binding and study of histidine-tagged proteins by surface plasmon resonance. Anal. Chem. 68, 490-497.
Spinke, J., Liley, M., Guder, H.J., Angermaier, L., Knoll, W., 1993. Molecular recognition at self-assembled monolayers: the construction of multicomponent multilayers. Langmuir 9 (7), 1821-1825.
Sun, X., He, P., Liu, S., Ye, Jiannog, Fang, Y., 1998. Immobilization of single- stranded deoxyribonucleic acid on gold electrode with self-assembled aminoethanethiol monolayer for DNA electrochemical sensor applications. Talanta 47, 487-495.
Wagner, P., Hegner, M., Guntherodt, H.J., Semenza, G., 1995. Formation and in situ modification of monolayers chemisorbed on ultraflat template-stripped gold surfaces. Langmuir 11, 3867-3875.
Wang, T.H., Chen Y.F., Masset S., Ho, CM., Tai, Y.C, 2000. Molecular beacon based micro biological detection system. Proceedings of the 2000 International Conference on Mathematics and Engineering Techniques in Medicine and Biological Sciences (METMBS'2000) Las Vegas, Nevada.
Wang, J., Rivas, G., Cai, X., Palecek, E., Nielsen, P., Shiraishi, H., Dontha, N., Luo, D., Parrado, C, Chicharro, M., Farias, P.A.M, Valera, F.S., Grant, D.H., Ozsoz, M., Flair, M.N., 1997. DNA electrochemical biosensors for environmental monitoring. A Review. Anal. Chim. Acta 347, 1-8. Weber, P.C., Ohlendorf, D.H., Wendoloski, J.J., Salemme, F.R., 1989. Structural
Origins of High- Affinity Biotin Binding to Streptavidin. Science 243, 85-88.
Xia, Y., Whitesides, G.M., 1998. Soft Lithography. Angew. Chem. Int. Ed. 37, 550-575. Table I
Comparison of various reagents for the from surface
Figure imgf000054_0001
Figure imgf000054_0005
Figure 1 Figure 2
Figure imgf000054_0002
2J8 αn
Figure 3
Figure imgf000054_0003
Figure .5 aitiitift
Figure imgf000054_0004
Figured
Figure imgf000055_0001
500 I ODD I50D 2000
Ηnuøecintdj)
Figure 7
Figure imgf000055_0002
()
Figure 8
Figure imgf000056_0001
Pctβrfia [Scan Rate]"1
Figure imgf000056_0002
Figure 10
Figure imgf000056_0003
Petødύ
Figure 11
Figure imgf000056_0004
lxEcαK O.lxE. coK Boidefella Figure 12
1000011
2,102
10000 738 £
1351
1000 7858
63
53 S 8
100
,V hV t ■ ' -V » 4 ^ v<* ^
Number of Cell;
Figure 13
Figure imgf000057_0001
lCHSEc i lOBEccB lOElEcdi ltlBEcdi 2ib tιB
BαrO-eHa
Number ofCells

Claims

Claims
1. A method of detecting the presence or measuring the quantity of a target analyte in a sample reagent comprising the steps of: contacting a microfabricated electrochemical biosensor with the sample reagent, the microfabricated electrochemical biosensor comprising:
(a) a substrate; and
(b) at least two electrically conductive electrodes fabricated on the substrate by integrated circuit technology, each of the electrical conductive electrodes consisting of a single layer of an electrically conductive material; containing the sample reagent in contact with the conducting electrodes; measuring a electrical signal output from the microfabricated electrochemical biosensor; and determining from the signal output the presence and/or quantity of the target analyte in the sample reagent.
2. The method of claim 1 wherein the electrochemical biosensor further comprises an adhesive underneath each of the electrodes, the adhesive allowing for better adhesion of each of the electrodes to the substrate.
3. The method of claim 2 wherein the sample reagent is a biological fluid containing macromolecules.
4. The method of claim 2 wherein the sample reagent is a biological fluid containing ionic molecules or atoms.
5. The method of claim 2 wherein the substrate is selected from the group consisting of silicon, gallium arsenide, plastic and glass.
6. The method of claim 2 wherein the substrate comprises a material made out of silicon.
7. The method of claim 2 wherein the electrically conductive material is selected from the group consisting of gold, aluminum, chromium, copper, platinum, titanium, nickel and titanium.
8. The method of claim 2 wherein the electrically conductive material is gold.
9. The method of claim 2 wherein the adhesive is selected from the group of consisting of chromium, titanium, and glue.
10. The method of claim 2 wherein the adhesive comprises chromium.
11. The method of claim 2 wherein the substrate further comprises a well structure containing at least one of the electrodes.
12. The method of claim 2 wherein the electrochemical biosensor comprises at least three electrically conductive electrodes.
13. The method of claim 12 wherein each of the electrically conductive electrodes consists of a single layer of gold.
14. The method of claim 2 wherein the step of determining from the signal output the presence and/or quantity of the target analyte in the reagent further comprises the steps of: calibrating the electrochemical biosensor with a first calibrating solution that contains a known amount of the target analyte to be detected and a second calibrating solution that contains an undetectable amount of the target analyte to be detected; obtaining a reference signal output; and comparing the reference signal with the measured signal to determine the presence and/or quantity of the molecules in the sample reagent.
15. The method of claim 14 wherein the substrate is selected from the group consisting of silicon, gallium arsenide, plastic and glass.
16. The method of claim 14 wherein the electrically conductive material is selected from the group consisting of gold, aluminum, chromium, copper, platinum, nickel and titanium.
17. The method of claim 14 wherein the electrically conductive material is gold.
18. The method of claim 14 wherein the adhesive is selected from the group of material consisting of chromium, titanium, and glue.
19. The method of claim 14 wherein the substrate further comprises a well structure underneath at least one of the electrodes.
20. The method of claim 14 wherein a surface on at least one of the electrodes is surface modified for anchoring macromolecules on the surface.
21. A method of detecting the presence or measuring the quantity of at least one molecule in a sample reagent comprising the steps of: contacting a microfabricated electrochemical biosensor with the sample reagent, the microfabricated electrochemical biosensor comprising: (a) a silicon substrate; and (b) three electrically conductive electrodes fabricated on the substrate by integrated circuit technology, each of the electrically conductive electrodes consisting of a single layer of gold; containing the sample reagent in contact with the conducting electrodes; measuring a signal output from the microfabricated electrochemical biosensor; and determining from the signal output the presence and/or quantity of the molecules in the sample reagent.
22. A microfabricated electrochemical biosensor comprising: a silicon substrate; and three electrical conductive electrodes for redox sensing fabricated on the substrate by integrated circuit technology, each of the electrical conductive electrodes consisting of a single layer of gold.
23. A method of detection of a target analyte comprising the steps of: providing a biosensor and at least one reagent, the biosensor comprising:
(a) a first area having a first surface property for immobilizing the target analyte contained in the reagent; and
(b) a second area having a second surface property different from the first surface property so that a sample of the reagent can be confined due to surface tension forces between the first area and the second area, the second area comprising components for detection of the target analyte; applying a volume of reagent to the biosensor, wherein the coverage of the reagent over the biosensor is controlled by the volume of the reagent and the surface tension forces; and detecting a presence and/or measuring a quantity of the target analyte by the biosensor.
24. The method of claim 23 wherein the components comprise at least one electrode selected from the group consisting of a counter electrode and a reference electrode.
25. The method of claim 23 wherein at least one reagent comprises a first reagent and a second reagent.
26. The method of claim 25 wherein the step of applying reagent to the biosensor comprises the steps of: covering the first reagent over the first area; and covering the second reagent over the second area.
27. The method of claim 26 wherein the first area is shaped in a first geometry that is designed to confine the first reagent within the first area.
28. The method of claim 27 wherein the second area is shaped in a second geometry that is designed to confine the second reagent with the second area.
29. The method of claim 25 wherein the first reagent is a biological fluid containing the target analyte and the second reagent is a chemical solution suitable for biological detection of the target analyte.
30. The method of claim 23 wherein the target analyte is selected from the group consisting of ionic molecule and macromolecule.
31. The method of claim 23 wherein the step of detecting the presence and/or quantity of the target analyte further comprises: calibrating the biosensor with a first calibrating solution that contains a known amount of the analyte to be detected and a second calibrating solution that contains an undetectable amount of the analyte to be detected; and utilizing the calibration results to detect the presence a quantity of target analyte in the reagent.
32. The method of claim 23 wherein the surface properties of the first and second areas are surface modified before application of reagent to the biosensor.
33. The method of claim 23 wherein the surface properties of the first and second areas are controlled by an external force field.
34. A device for detecting a redox event of at least one analyte in a liquid reagent comprising: a redox sensor fabricated on a semiconductor by using at least one of integrated circuit (IC) and micro electromechanical systems (MEMS) technology, the redox sensor includes at least two electrically conductive electrodes for signal output and bias control; an isolation layer between the electrodes and the semiconductor; an integrated circuit (IC) on the semiconductor and underneath the isolation layer, the integrated circuit comprising a detection circuit and a bias potential circuit for providing a bias potential to the detection circuit; and wherein the isolation layer has a electrically conductive contact portion for electrically connecting the electrodes with the integrated circuit (IC).
35. The device of claim 34 wherein the electrically conductive electrodes are selected from the group of materials consisting of gold, aluminum, chromium, copper, platinum, nickel and titanium.
36. The device of claim 34 wherein the electrically conductive electrodes are made out of gold.
37. The device of claim 34 wherein the semiconductor is selected from the group consisting of silicon and gallium arsenide.
38. The device of claim 34 wherein the semiconductor is silicon.
39. The device of claim 34 wherein the detection circuit comprises a current measurement device to detect a current signal from the redox event through a corresponding electrically connected electrode.
40. The device of claim 39 wherein the bias potential is a potential difference between at least two different electrodes to enable electron transfer for current detection.
41. A device for detecting a redox event of at least one analyte in a liquid reagent comprising: a redox sensor on a semiconductor, the redox sensor comprising a reference electrode, a working electrode and a counter electrode; an isolation layer between the redox sensor and the semiconductor; an integrated circuit (IC) on the semiconductor and underneath the isolation layer, the integrated circuit comprising a detection circuit and a bias potential circuit for providing a bias potential to the detection circuit; and wherein the isolation layer has a electrically conductive contact portion for electrically connecting the electrodes with the integrated circuit (IC).
42. The device of claim 41 wherein each of the electrodes consists of a single layer of electrically conductive material.
43. The device of claim 42 wherein the electrically conductive material is gold.
44. The device of claim 43 wherein the semiconductor is silicon.
45. The device of claim 44 wherein the detection circuit comprises a current measurement device to detect a current signal from the redox event through a corresponding electrically conductive electrode.
46. The device of claim 45 wherein the bias potential is a potential difference between at least two different electrodes to enable electron transfer for current detection.
47. The device of claim 41 wherein the semiconductor is selected from the group consisting of silicon and gallium arsenide.
48. The device of claim 41 wherein the semiconductor is silicon.
49. The device of claim 41 wherein the detection circuit comprises a current measurement device to detect a current signal from the redox event through a corresponding electrically conductive electrode.
50. The device of claim 49 wherein the bias potential is a potential difference between at least two different electrodes to enable electron transfer for current detection.
PCT/US2001/014257 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip WO2001083674A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA2407973A CA2407973C (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip
AU6114501A AU6114501A (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip
JP2001580284A JP4949589B2 (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip
AU2001261145A AU2001261145B2 (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip
EP01935016A EP1278821A4 (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20160300P 2000-05-03 2000-05-03
US60/201,603 2000-05-03

Publications (1)

Publication Number Publication Date
WO2001083674A1 true WO2001083674A1 (en) 2001-11-08

Family

ID=22746508

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/014257 WO2001083674A1 (en) 2000-05-03 2001-05-02 Biological identification system with integrated sensor chip

Country Status (8)

Country Link
US (2) US7399585B2 (en)
EP (1) EP1278821A4 (en)
JP (1) JP4949589B2 (en)
CN (1) CN100457887C (en)
AU (2) AU6114501A (en)
CA (1) CA2407973C (en)
TW (1) TWI245073B (en)
WO (1) WO2001083674A1 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002058846A3 (en) * 2001-01-24 2003-04-24 Univ Michigan Micromachined device for receiving and retaining at least one liquid droplet, method of making the device and method of using the device
WO2003079016A1 (en) * 2002-03-18 2003-09-25 Infineon Technologies Ag sIOSENSOR FOR DETECTING MACROMOLECULAR BIOPOLYMERS AND METHOD FOR THE PRODUCTION THEREOF
KR20030075359A (en) * 2002-03-18 2003-09-26 학교법인 포항공과대학교 Multiplex nucleic acid amplification device using micro electro mechanical system component and method for constructing same
WO2003083134A1 (en) * 2002-04-03 2003-10-09 Infineon Technologies Ag Sensor for the quantitative and qualitative determination of (bio)organic oligomers and polymers, corresponding analysis method, and method for the production of said sensor
JP2004532396A (en) * 2001-03-09 2004-10-21 シーメンス アクチエンゲゼルシヤフト Module for analyzer, applicator and analyzer as replacement part of analyzer
WO2004092712A1 (en) * 2003-04-18 2004-10-28 Hitachi Chemical Co., Ltd. Molecule detecting method, molecule counting method, molecule localization detecting method, molecule detection device used for them
EP1497458A2 (en) * 2002-04-10 2005-01-19 Geneohm Sciences, Inc. Hydrophobic zone device
WO2005106034A1 (en) 2004-04-29 2005-11-10 Agency For Science, Technology And Research Method and device for detection of nucleic acids and/or polypeptides
JP2005534938A (en) * 2002-08-06 2005-11-17 ザ・リージェンツ・オブ・ザ・ユニバーシティー・オブ・カリフォルニア Tear membrane osmotic pressure method
WO2005111597A1 (en) * 2004-05-17 2005-11-24 The Circle For The Promotion Of Science And Engineering Sensor for magnetic fine particle
AT500427A1 (en) * 2002-05-29 2005-12-15 Ronacher Bernhard Mag Dr DEVICE FOR ANALYZING INGREDIENTS OF A SAMPLE
WO2005059513A3 (en) * 2003-12-15 2006-05-26 Geneohm Sciences Inc Multiplexed electrochemical detection system and method
WO2006133042A2 (en) * 2005-06-06 2006-12-14 Intel Corporation Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
US7153687B2 (en) 2002-08-13 2006-12-26 Hong Kong Dna Chips Limited Apparatus and methods for detecting DNA in biological samples
US7572624B2 (en) * 2002-12-19 2009-08-11 Siemens Aktiengesellschaft DNA chip comprising a microarray made of an microelectrode system
WO2013017635A1 (en) * 2011-08-04 2013-02-07 Universität Rostock Electrochemical sensor
WO2013058646A1 (en) * 2011-10-19 2013-04-25 Universiti Sains Malaysia A system for identifying presence of polynucleotides of interest from a sample
US8470144B2 (en) 2010-12-28 2013-06-25 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US8691062B2 (en) 2010-12-28 2014-04-08 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US8691061B2 (en) 2010-12-28 2014-04-08 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US8940143B2 (en) 2007-06-29 2015-01-27 Intel Corporation Gel-based bio chip for electrochemical synthesis and electrical detection of polymers
TWI486586B (en) * 2013-01-16 2015-06-01 Univ Nat Chi Nan Current - type biological sensor and its making method
RU2570706C1 (en) * 2014-07-22 2015-12-10 Федеральное государственное унитарное предприятие "Государственный научный центр "Научно-исследовательский институт органических полупродуктов и красителей" (ФГУП "ГНЦ "НИОПИК") Method for qantitative determination of sodium fluorescein in substance and thereof-based medication
US9335243B2 (en) 2006-12-11 2016-05-10 Tearlab Research, Inc. Systems and methods for collecting tear film and measuring tear film osmolarity
EP3260853A1 (en) * 2009-03-27 2017-12-27 ams International AG A sensor device and a method of manufacturing the same
DE102021200588A1 (en) 2021-01-22 2022-07-28 Robert Bosch Gesellschaft mit beschränkter Haftung Method and control device for producing a carrier element for receiving a sample liquid, carrier element and analysis device with carrier element
US11536707B2 (en) 2014-09-23 2022-12-27 Tearlab Research, Inc. Systems and methods for integration of microfluidic tear collection and lateral flow analysis of analytes of interest
US11815430B2 (en) 2019-02-22 2023-11-14 Hewlett-Packard Development Company, L.P. Nucleic acid detection

Families Citing this family (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2407973C (en) 2000-05-03 2011-06-07 Jen-Jr Gau Biological identification system with integrated sensor chip
US8329010B2 (en) * 2000-05-03 2012-12-11 Kotura, Inc. Chip assay having improved efficiency
AU2002241803A1 (en) 2000-10-20 2002-06-18 The Board Of Trustees Of The Leland Stanford Junior University Transient electrical signal based methods and devices for characterizing molecular interaction and/or motion in a sample
WO2002103037A1 (en) * 2001-06-19 2002-12-27 Molecular Circuitry, Inc. Conductometric detection process
DE60220804D1 (en) * 2001-08-20 2007-08-02 Regenesis Bioremediation Produ BIOSENSOR FOR SMALL MOLECULAR ANALYTES
US20070178477A1 (en) * 2002-01-16 2007-08-02 Nanomix, Inc. Nanotube sensor devices for DNA detection
US20030134433A1 (en) * 2002-01-16 2003-07-17 Nanomix, Inc. Electronic sensing of chemical and biological agents using functionalized nanostructures
US20060228723A1 (en) * 2002-01-16 2006-10-12 Keith Bradley System and method for electronic sensing of biomolecules
US8010174B2 (en) 2003-08-22 2011-08-30 Dexcom, Inc. Systems and methods for replacing signal artifacts in a glucose sensor data stream
US8260393B2 (en) 2003-07-25 2012-09-04 Dexcom, Inc. Systems and methods for replacing signal data artifacts in a glucose sensor data stream
US7948041B2 (en) * 2005-05-19 2011-05-24 Nanomix, Inc. Sensor having a thin-film inhibition layer
US20040005572A1 (en) * 2002-07-05 2004-01-08 Rosner S. Jeffrey Electronically readable microarrays
US20040038385A1 (en) * 2002-08-26 2004-02-26 Langlois Richard G. System for autonomous monitoring of bioagents
DE10243569A1 (en) * 2002-09-19 2004-04-01 Infineon Technologies Ag Circuit arrangement and method for producing a circuit arrangement
US20070099194A1 (en) * 2005-11-02 2007-05-03 Xing Yang Quality control of probe attachment on electrodes
US8282549B2 (en) 2003-12-09 2012-10-09 Dexcom, Inc. Signal processing for continuous analyte sensor
US8160669B2 (en) 2003-08-01 2012-04-17 Dexcom, Inc. Transcutaneous analyte sensor
US7591801B2 (en) 2004-02-26 2009-09-22 Dexcom, Inc. Integrated delivery device for continuous glucose sensor
US20190357827A1 (en) 2003-08-01 2019-11-28 Dexcom, Inc. Analyte sensor
US8275437B2 (en) 2003-08-01 2012-09-25 Dexcom, Inc. Transcutaneous analyte sensor
US20140121989A1 (en) 2003-08-22 2014-05-01 Dexcom, Inc. Systems and methods for processing analyte sensor data
US7920906B2 (en) 2005-03-10 2011-04-05 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US9247900B2 (en) 2004-07-13 2016-02-02 Dexcom, Inc. Analyte sensor
US20050112587A1 (en) * 2003-11-25 2005-05-26 Sherrill James V. Analyzing biological probes
US8364231B2 (en) 2006-10-04 2013-01-29 Dexcom, Inc. Analyte sensor
US8774886B2 (en) 2006-10-04 2014-07-08 Dexcom, Inc. Analyte sensor
US8423114B2 (en) 2006-10-04 2013-04-16 Dexcom, Inc. Dual electrode system for a continuous analyte sensor
EP2239567B1 (en) 2003-12-05 2015-09-02 DexCom, Inc. Calibration techniques for a continuous analyte sensor
US11633133B2 (en) 2003-12-05 2023-04-25 Dexcom, Inc. Dual electrode system for a continuous analyte sensor
DE602004028164D1 (en) * 2003-12-08 2010-08-26 Dexcom Inc SYSTEMS AND METHOD FOR IMPROVING ELECTROCHEMICAL ANALYTIC SENSORS
CN1906649A (en) * 2003-12-10 2007-01-31 史密斯探测公司 Autonomous surveillance system
US8808228B2 (en) 2004-02-26 2014-08-19 Dexcom, Inc. Integrated medicament delivery device for use with continuous analyte sensor
US7217428B2 (en) 2004-05-28 2007-05-15 Technology Innovations Llc Drug delivery apparatus utilizing cantilever
JP4143046B2 (en) * 2004-06-02 2008-09-03 株式会社東芝 Nucleic acid detection substrate and nucleic acid detection method using the apparatus
US8565848B2 (en) 2004-07-13 2013-10-22 Dexcom, Inc. Transcutaneous analyte sensor
US8452368B2 (en) 2004-07-13 2013-05-28 Dexcom, Inc. Transcutaneous analyte sensor
US20060020192A1 (en) 2004-07-13 2006-01-26 Dexcom, Inc. Transcutaneous analyte sensor
US7640048B2 (en) 2004-07-13 2009-12-29 Dexcom, Inc. Analyte sensor
US7763426B2 (en) * 2004-11-01 2010-07-27 The Regents Of The University Of California Probes and methods for detection of Escheridia coli and antibiotic resistance
WO2007008246A2 (en) * 2004-11-12 2007-01-18 The Board Of Trustees Of The Leland Stanford Junior University Charge perturbation detection system for dna and other molecules
US20090178937A1 (en) * 2004-12-29 2009-07-16 David William Taylor Analyte measurement meter or system incorporating an improved measurement circuit
WO2006096761A1 (en) * 2005-03-08 2006-09-14 Authentix, Inc. Microfluidic device for identification, quantification, and authentication of latent markers
WO2006116455A2 (en) 2005-04-26 2006-11-02 Applera Corporation System for genetic surveillance and analysis
US20070292855A1 (en) * 2005-08-19 2007-12-20 Intel Corporation Method and CMOS-based device to analyze molecules and nanomaterials based on the electrical readout of specific binding events on functionalized electrodes
US8880138B2 (en) 2005-09-30 2014-11-04 Abbott Diabetes Care Inc. Device for channeling fluid and methods of use
JP4735833B2 (en) * 2006-01-13 2011-07-27 セイコーエプソン株式会社 Biochip and biosensor
US7826879B2 (en) 2006-02-28 2010-11-02 Abbott Diabetes Care Inc. Analyte sensors and methods of use
US11001881B2 (en) 2006-08-24 2021-05-11 California Institute Of Technology Methods for detecting analytes
US11525156B2 (en) 2006-07-28 2022-12-13 California Institute Of Technology Multiplex Q-PCR arrays
US8048626B2 (en) 2006-07-28 2011-11-01 California Institute Of Technology Multiplex Q-PCR arrays
US11560588B2 (en) 2006-08-24 2023-01-24 California Institute Of Technology Multiplex Q-PCR arrays
JP2010503856A (en) * 2006-09-14 2010-02-04 エージェンシー フォー サイエンス, テクノロジー アンド リサーチ Electrochemical sensor with comb-shaped microelectrode and conductive polymer
US7932034B2 (en) 2006-12-20 2011-04-26 The Board Of Trustees Of The Leland Stanford Junior University Heat and pH measurement for sequencing of DNA
WO2008102120A1 (en) * 2007-02-20 2008-08-28 Oxford Nanopore Technologies Limited Lipid bilayer sensor system
GB2447043A (en) * 2007-02-20 2008-09-03 Oxford Nanolabs Ltd Lipid bilayer sensor system
EP2152350A4 (en) 2007-06-08 2013-03-27 Dexcom Inc Integrated medicament delivery device for use with continuous analyte sensor
US20090024015A1 (en) * 2007-07-17 2009-01-22 Edwards Lifesciences Corporation Sensing element having an adhesive backing
EP4098177A1 (en) 2007-10-09 2022-12-07 DexCom, Inc. Integrated insulin delivery system with continuous glucose sensor
US8051697B2 (en) * 2007-10-17 2011-11-08 The George Washington University Self calibration devices for chemical and bio analytical trace detection systems
US20090155948A1 (en) * 2007-12-18 2009-06-18 National Applied Research Laboratories Methods for manufacturing cmos compatible bio-sensors
GB0724736D0 (en) 2007-12-19 2008-01-30 Oxford Nanolabs Ltd Formation of layers of amphiphilic molecules
CA2715628A1 (en) 2008-02-21 2009-08-27 Dexcom, Inc. Systems and methods for processing, transmitting and displaying sensor data
KR101046720B1 (en) * 2008-06-30 2011-07-05 주식회사 하이닉스반도체 Molecular electronic device and manufacturing method thereof
TR200806315A2 (en) * 2008-08-22 2010-03-22 Külah Haluk Concentric electrode and spiral microfluidic channel dielectrophoretic microcell chromatography device manufactured with MEMS technology
JP2010071604A (en) * 2008-09-22 2010-04-02 Mitsubishi Electric Corp Humidifying device
US9709560B2 (en) * 2008-09-29 2017-07-18 Intel Corporation Biosensors and biosensing incorporating RF and microwave radiation
JP2011018885A (en) * 2009-06-12 2011-01-27 Seiko Epson Corp Method of manufacturing patterned film forming member, patterned film forming member, electro-optical device, electronic apparatus
DE102009043527B4 (en) * 2009-09-30 2021-06-10 Boehringer Ingelheim Vetmedica Gmbh Arrangement and method using microsensors for measuring cell vitalities
WO2012142502A2 (en) 2011-04-15 2012-10-18 Dexcom Inc. Advanced analyte sensor calibration and error detection
US8546080B2 (en) * 2011-06-17 2013-10-01 International Business Machines Corporation Molecular dispensers
DE102011108885A1 (en) * 2011-07-28 2013-01-31 Forschungszentrum Jülich GmbH Electrode arrangement and method for operating the electrode arrangement
US9117610B2 (en) 2011-11-30 2015-08-25 General Electric Company Integrated micro-electromechanical switches and a related method thereof
GB201202519D0 (en) 2012-02-13 2012-03-28 Oxford Nanopore Tech Ltd Apparatus for supporting an array of layers of amphiphilic molecules and method of forming an array of layers of amphiphilic molecules
WO2013165325A1 (en) 2012-05-04 2013-11-07 Haluk Kulah Micro electrochemical sensor
DK2844762T3 (en) 2012-05-04 2018-03-26 Univ California TESTING FOR ANTIBIOTIC SENSITIVITY USING PROBRIOSOMAL RNA PROBLEMS
WO2013176773A1 (en) 2012-05-24 2013-11-28 The Governing Council Of The University Of Toronto Systems and methods for multiplexed electrochemical detection
GB201313121D0 (en) 2013-07-23 2013-09-04 Oxford Nanopore Tech Ltd Array of volumes of polar medium
US9623409B2 (en) 2013-03-11 2017-04-18 Cue Inc. Cartridges, kits, and methods for enhanced mixing for detection and quantification of analytes
US10545161B2 (en) 2013-03-11 2020-01-28 Cue Health Inc. Systems and methods for detection and quantification of analytes
EP2972333B1 (en) 2013-03-11 2018-09-19 The University of Toledo A biosensor device to target analytes in situ, in vivo, and/or in real time, and methods of making and using the same
CA3160098A1 (en) 2013-03-11 2014-10-09 Cue Health Inc. Systems and methods for detection and quantification of analytes
WO2014152717A2 (en) 2013-03-14 2014-09-25 Sano Intelligence, Inc. On-body microsensor for biomonitoring
US10820860B2 (en) 2013-03-14 2020-11-03 One Drop Biosensor Technologies, Llc On-body microsensor for biomonitoring
US9983163B2 (en) 2013-04-30 2018-05-29 Board Of Regents, The University Of Texas System Integrated electro-analytical biosensor array
DE102013210138A1 (en) * 2013-05-30 2014-12-04 Boehringer Ingelheim Vetmedica Gmbh Method for generating a plurality of measuring ranges on a chip and chip with measuring ranges
CN106102578A (en) 2014-03-13 2016-11-09 萨诺智能公司 For monitoring the system of body chemistry
US10595754B2 (en) 2014-03-13 2020-03-24 Sano Intelligence, Inc. System for monitoring body chemistry
USD745423S1 (en) 2014-05-12 2015-12-15 Cue Inc. Automated analyzer test cartridge and sample collection device for analyte detection
CN106999929A (en) * 2014-09-11 2017-08-01 克忧公司 For detecting the system and method with analyte quantification
GB201418512D0 (en) 2014-10-17 2014-12-03 Oxford Nanopore Tech Ltd Electrical device with detachable components
US10473638B2 (en) 2014-12-19 2019-11-12 Siemens Healthcare Diagnostics Inc. Methods and systems for improving precision of measurements for reduced sample volumes
EP3314245A4 (en) 2015-06-25 2019-02-27 Roswell Biotechnologies, Inc Biomolecular sensors and methods
CN112881730A (en) 2015-07-17 2021-06-01 克忧健康公司 Systems and methods for enhanced detection and analyte quantitation
CN105137023B (en) * 2015-10-14 2018-08-03 浙江公正检验中心有限公司 A kind of System and method for of on-line checking aquatic products
JP6116075B1 (en) 2015-11-20 2017-04-19 日本航空電子工業株式会社 Electrochemical measurement method, electrochemical measurement apparatus and transducer
CA3003767A1 (en) 2015-12-02 2017-06-08 Boehringer Ingelheim Vetmedica Gmbh Method for producing a plurality of measurement regions on a chip, and chip having a plurality of measurement regions
TWI656345B (en) * 2016-01-13 2019-04-11 Prostate cancer detection module and its operation method
WO2017132586A1 (en) 2016-01-28 2017-08-03 Roswell Biotechnologies, Inc. Methods and apparatus for measuring analytes using large scale molecular electronics sensor arrays
US10712334B2 (en) 2016-01-28 2020-07-14 Roswell Biotechnologies, Inc. Massively parallel DNA sequencing apparatus
US10737263B2 (en) 2016-02-09 2020-08-11 Roswell Biotechnologies, Inc. Electronic label-free DNA and genome sequencing
US10597767B2 (en) 2016-02-22 2020-03-24 Roswell Biotechnologies, Inc. Nanoparticle fabrication
WO2017155858A1 (en) 2016-03-07 2017-09-14 Insilixa, Inc. Nucleic acid sequence identification using solid-phase cyclic single base extension
WO2016176692A2 (en) * 2016-06-17 2016-11-03 Ohmx Corporation Biosensor chip
GB201611770D0 (en) 2016-07-06 2016-08-17 Oxford Nanopore Tech Microfluidic device
WO2018022026A1 (en) 2016-07-26 2018-02-01 Hewlett-Packard Development Company, L.P. Microfluidic apparatuses for fluid movement control
US9829456B1 (en) 2016-07-26 2017-11-28 Roswell Biotechnologies, Inc. Method of making a multi-electrode structure usable in molecular sensing devices
US11268927B2 (en) 2016-08-30 2022-03-08 Analog Devices International Unlimited Company Electrochemical sensor, and a method of forming an electrochemical sensor
US10620151B2 (en) * 2016-08-30 2020-04-14 Analog Devices Global Electrochemical sensor, and a method of forming an electrochemical sensor
JP6218199B1 (en) * 2016-10-06 2017-10-25 日本航空電子工業株式会社 Electrochemical measuring device and transducer
EP3568407A4 (en) 2017-01-10 2020-12-23 Roswell Biotechnologies, Inc Methods and systems for dna data storage
EP3571286A4 (en) 2017-01-19 2020-10-28 Roswell Biotechnologies, Inc Solid state sequencing devices comprising two dimensional layer materials
WO2018140540A1 (en) * 2017-01-25 2018-08-02 Cue Health Inc. Systems and methods for enhanced detection and quantification of analytes
US10508296B2 (en) 2017-04-25 2019-12-17 Roswell Biotechnologies, Inc. Enzymatic circuits for molecular sensors
EP3615685A4 (en) 2017-04-25 2021-01-20 Roswell Biotechnologies, Inc Enzymatic circuits for molecular sensors
EP3622086A4 (en) 2017-05-09 2021-04-21 Roswell Biotechnologies, Inc Binding probe circuits for molecular sensors
KR101921627B1 (en) * 2017-06-16 2018-11-26 한국과학기술연구원 Field effect transistor, biosensor comprising the same, method for manufacturing Field effect transistor, and method for manufacturing biosensor
KR20200039795A (en) 2017-08-30 2020-04-16 로스웰 바이오테크놀로지스 인코포레이티드 Progressive enzyme molecular electronic sensors for DNA data storage
KR20200067871A (en) 2017-10-10 2020-06-12 로스웰 바이오테크놀로지스 인코포레이티드 Methods, devices and systems for storing amplified DNA data
US11331022B2 (en) 2017-10-24 2022-05-17 Dexcom, Inc. Pre-connected analyte sensors
US11382540B2 (en) 2017-10-24 2022-07-12 Dexcom, Inc. Pre-connected analyte sensors
US11022579B2 (en) 2018-02-05 2021-06-01 Analog Devices International Unlimited Company Retaining cap
KR102328745B1 (en) * 2018-04-17 2021-11-22 한국화학연구원 Multi-well electrode based biosensor
GB2573323A (en) 2018-05-03 2019-11-06 Mursia Ltd Biosensor method and system
US11002730B2 (en) 2018-05-23 2021-05-11 International Business Machines Corporation Molecular design to suppress desorption of self-assembled monolayers
CN110609064A (en) * 2018-06-14 2019-12-24 深圳碳森科技有限公司 Differential impedance potential type biosensor and manufacturing method thereof
US10738342B2 (en) 2018-08-30 2020-08-11 Urinary Technologies, Inc. System for microbial species detection, quantification and antibiotic susceptibility identification
CN109504603A (en) * 2018-10-25 2019-03-22 江苏省产品质量监督检验研究院 The detection method of total plate count in a kind of food
CN109307541B (en) * 2018-11-29 2020-03-24 郑州安图生物工程股份有限公司 Method for measuring volume of lighting plate single-hole residual liquid
WO2020123957A2 (en) * 2018-12-14 2020-06-18 Cepheid Diagnostic detection chip devices and methods of manufacture and assembly
US11406299B2 (en) * 2019-02-22 2022-08-09 International Business Machines Corporation Biosensors with programmable sensing cavities
EP3938779A1 (en) 2019-03-12 2022-01-19 Oxford Nanopore Technologies Limited Nanopore sensing device and methods of operation and of forming it
EP3937780A4 (en) 2019-03-14 2022-12-07 InSilixa, Inc. Methods and systems for time-gated fluorescent-based detection
TWI708057B (en) * 2019-06-05 2020-10-21 王錦弘 Centrifugal reaction microtube, centrifugal reaction device and centrifugal inspection method thereof
TWI736928B (en) * 2019-07-10 2021-08-21 昇陽國際半導體股份有限公司 Method of depositing a gold film on a silicon wafer
TWI722484B (en) * 2019-07-10 2021-03-21 昇陽國際半導體股份有限公司 Electrochemical sensor with three-dimensional structure electrodes
CN117783242A (en) * 2019-08-20 2024-03-29 深圳硅基传感科技有限公司 Working electrode of glucose sensor suitable for mass production
US20210122926A1 (en) * 2019-10-29 2021-04-29 Nanoxcoatings Lc Protection of surfaces by evaporated salt coatings
US11060995B1 (en) * 2020-07-20 2021-07-13 Texas Tech University System Rapid viral diagnostic sensor
TWI781587B (en) * 2021-04-14 2022-10-21 財團法人金屬工業研究發展中心 Sensing electrode
USD988882S1 (en) 2021-04-21 2023-06-13 Informed Data Systems Inc. Sensor assembly
US11513097B1 (en) 2021-05-21 2022-11-29 PERSOWN, Inc. Methods of obtaining and using electrochemical diagnostic results
CN114324488B (en) * 2021-11-25 2023-05-16 中国科学院海洋研究所 Friction nano-generator driven sensing system and method for bacteria detection
CN114235921A (en) * 2022-02-23 2022-03-25 捷仪科技(北京)有限公司 Electrode slide glass for biological detection

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4225410A (en) * 1978-12-04 1980-09-30 Technicon Instruments Corporation Integrated array of electrochemical sensors
US5313264A (en) * 1988-11-10 1994-05-17 Pharmacia Biosensor Ab Optical biosensor system
US5466348A (en) * 1991-10-21 1995-11-14 Holm-Kennedy; James W. Methods and devices for enhanced biochemical sensing
US5766934A (en) * 1989-03-13 1998-06-16 Guiseppi-Elie; Anthony Chemical and biological sensors having electroactive polymer thin films attached to microfabricated devices and possessing immobilized indicator moieties
US5849486A (en) * 1993-11-01 1998-12-15 Nanogen, Inc. Methods for hybridization analysis utilizing electrically controlled hybridization
GB2335278A (en) * 1998-03-13 1999-09-15 Cygnus Therapeutic Systems Biosensor with reverse iontophoretic sampling system
US5981268A (en) * 1997-05-30 1999-11-09 Board Of Trustees, Leland Stanford, Jr. University Hybrid biosensors
US6251595B1 (en) * 1998-06-18 2001-06-26 Agilent Technologies, Inc. Methods and devices for carrying out chemical reactions

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4963245A (en) * 1986-05-02 1990-10-16 Ciba Corning Diagnostics Corp. Unitary multiple electrode sensor
US4900405A (en) * 1987-07-15 1990-02-13 Sri International Surface type microelectronic gas and vapor sensor
JP2536780B2 (en) * 1988-11-10 1996-09-18 株式会社エー・アンド・デイ Pick-up type enzyme electrode
US5200051A (en) * 1988-11-14 1993-04-06 I-Stat Corporation Wholly microfabricated biosensors and process for the manufacture and use thereof
JPH04118554A (en) * 1989-12-28 1992-04-20 Tosoh Corp Electrochemical method for measuring enzyme and bio-sensor
US5108819A (en) * 1990-02-14 1992-04-28 Eli Lilly And Company Thin film electrical component
US5120421A (en) * 1990-08-31 1992-06-09 The United States Of America As Represented By The United States Department Of Energy Electrochemical sensor/detector system and method
US6017696A (en) * 1993-11-01 2000-01-25 Nanogen, Inc. Methods for electronic stringency control for molecular biological analysis and diagnostics
US5632957A (en) * 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
DE4236421A1 (en) * 1992-10-28 1994-05-11 Horst Dr Ing Habil Ahlers Sensor arrangement, esp. bio and=or chemical sensor - has field elements on substrate with electrodes of varying shape and size
JP3500772B2 (en) * 1994-06-30 2004-02-23 Nok株式会社 Protein biosensor and measurement method using the same
DE4430023A1 (en) 1994-08-24 1996-02-29 Boehringer Mannheim Gmbh Electrochemical sensor
JPH08327584A (en) * 1995-06-02 1996-12-13 Tokuyama Corp Ion-selective electrode and method for measuring ion concentration
US5567302A (en) * 1995-06-07 1996-10-22 Molecular Devices Corporation Electrochemical system for rapid detection of biochemical agents that catalyze a redox potential change
AUPN661995A0 (en) 1995-11-16 1995-12-07 Memtec America Corporation Electrochemical cell 2
NL1004319C2 (en) * 1996-10-18 1998-04-21 Pelleting Technologie Nederlan Pelleteer.
JP3375040B2 (en) * 1997-07-29 2003-02-10 松下電器産業株式会社 Substrate quantification method
AU9599498A (en) * 1997-09-30 1999-04-23 M-Biotech, Inc. Biosensor
WO1999062919A1 (en) * 1998-06-01 1999-12-09 Roche Diagnostics Corporation Redox reversible bipyridyl osmium complex conjugates
US6290839B1 (en) * 1998-06-23 2001-09-18 Clinical Micro Sensors, Inc. Systems for electrophoretic transport and detection of analytes
DE19916921A1 (en) * 1999-04-14 2000-10-19 Fraunhofer Ges Forschung Electrical sensor array
US6518024B2 (en) * 1999-12-13 2003-02-11 Motorola, Inc. Electrochemical detection of single base extension
CA2407973C (en) 2000-05-03 2011-06-07 Jen-Jr Gau Biological identification system with integrated sensor chip
US7767437B2 (en) 2001-11-02 2010-08-03 Genefluidics, Inc. System for detection of a component in a liquid
GB0130318D0 (en) 2001-12-19 2002-02-06 Univ Leeds Membrane
CA2480770A1 (en) 2002-04-03 2003-10-09 Japan Science And Technology Agency Biochip sensor surface carrying polyethylene glycolated nanoparticles
US20060160100A1 (en) 2005-01-19 2006-07-20 Agency For Science, Technology And Research Enzymatic electrochemical detection assay using protective monolayer and device therefor

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4225410A (en) * 1978-12-04 1980-09-30 Technicon Instruments Corporation Integrated array of electrochemical sensors
US5313264A (en) * 1988-11-10 1994-05-17 Pharmacia Biosensor Ab Optical biosensor system
US5766934A (en) * 1989-03-13 1998-06-16 Guiseppi-Elie; Anthony Chemical and biological sensors having electroactive polymer thin films attached to microfabricated devices and possessing immobilized indicator moieties
US5466348A (en) * 1991-10-21 1995-11-14 Holm-Kennedy; James W. Methods and devices for enhanced biochemical sensing
US5849486A (en) * 1993-11-01 1998-12-15 Nanogen, Inc. Methods for hybridization analysis utilizing electrically controlled hybridization
US5981268A (en) * 1997-05-30 1999-11-09 Board Of Trustees, Leland Stanford, Jr. University Hybrid biosensors
GB2335278A (en) * 1998-03-13 1999-09-15 Cygnus Therapeutic Systems Biosensor with reverse iontophoretic sampling system
US6251595B1 (en) * 1998-06-18 2001-06-26 Agilent Technologies, Inc. Methods and devices for carrying out chemical reactions

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002058846A3 (en) * 2001-01-24 2003-04-24 Univ Michigan Micromachined device for receiving and retaining at least one liquid droplet, method of making the device and method of using the device
US6764652B2 (en) 2001-01-24 2004-07-20 The Regents Of The University Of Michigan Micromachined device for receiving and retaining at least one liquid droplet, method of making the device and method of using the device
US7438851B2 (en) 2001-01-24 2008-10-21 The Regents Of The University Of Michigan Microsensor with a well having a membrane disposed therein
JP2004532396A (en) * 2001-03-09 2004-10-21 シーメンス アクチエンゲゼルシヤフト Module for analyzer, applicator and analyzer as replacement part of analyzer
WO2003079016A1 (en) * 2002-03-18 2003-09-25 Infineon Technologies Ag sIOSENSOR FOR DETECTING MACROMOLECULAR BIOPOLYMERS AND METHOD FOR THE PRODUCTION THEREOF
KR20030075359A (en) * 2002-03-18 2003-09-26 학교법인 포항공과대학교 Multiplex nucleic acid amplification device using micro electro mechanical system component and method for constructing same
WO2003083134A1 (en) * 2002-04-03 2003-10-09 Infineon Technologies Ag Sensor for the quantitative and qualitative determination of (bio)organic oligomers and polymers, corresponding analysis method, and method for the production of said sensor
EP1497458A2 (en) * 2002-04-10 2005-01-19 Geneohm Sciences, Inc. Hydrophobic zone device
EP1497458A4 (en) * 2002-04-10 2005-05-11 Geneohm Sciences Inc Hydrophobic zone device
AT500427B1 (en) * 2002-05-29 2009-02-15 Anagnostics Bioanalysis Gmbh DEVICE FOR ANALYZING INGREDIENTS OF A SAMPLE
AT500427A1 (en) * 2002-05-29 2005-12-15 Ronacher Bernhard Mag Dr DEVICE FOR ANALYZING INGREDIENTS OF A SAMPLE
JP2005534938A (en) * 2002-08-06 2005-11-17 ザ・リージェンツ・オブ・ザ・ユニバーシティー・オブ・カリフォルニア Tear membrane osmotic pressure method
US8110669B2 (en) 2002-08-13 2012-02-07 Hai Kang Life Corporation Limited Apparatus and methods for detecting DNA in biological samples
US7153687B2 (en) 2002-08-13 2006-12-26 Hong Kong Dna Chips Limited Apparatus and methods for detecting DNA in biological samples
US8198070B2 (en) 2002-08-13 2012-06-12 Hai Kang Life Corporation Limited Apparatus and methods for detecting DNA in biological samples
US7749705B2 (en) 2002-08-13 2010-07-06 Hai Kang Life Corporation Limited Apparatus and methods for detecting DNA in biological samples
US7572624B2 (en) * 2002-12-19 2009-08-11 Siemens Aktiengesellschaft DNA chip comprising a microarray made of an microelectrode system
WO2004092712A1 (en) * 2003-04-18 2004-10-28 Hitachi Chemical Co., Ltd. Molecule detecting method, molecule counting method, molecule localization detecting method, molecule detection device used for them
WO2005059513A3 (en) * 2003-12-15 2006-05-26 Geneohm Sciences Inc Multiplexed electrochemical detection system and method
US8420313B2 (en) 2003-12-15 2013-04-16 Geneohm Sciences, Inc. Multiplexed electrochemical detection system and method
US7341834B2 (en) 2003-12-15 2008-03-11 Geneohn Sciences, Inc. Multiplexed electrochemical detection system and method
AU2004300204B2 (en) * 2003-12-15 2010-06-24 Geneohm Science, Inc. Multiplexed electrochemical detection system and method
WO2005106034A1 (en) 2004-04-29 2005-11-10 Agency For Science, Technology And Research Method and device for detection of nucleic acids and/or polypeptides
WO2005111597A1 (en) * 2004-05-17 2005-11-24 The Circle For The Promotion Of Science And Engineering Sensor for magnetic fine particle
WO2006133042A3 (en) * 2005-06-06 2007-02-01 Intel Corp Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
US8278121B2 (en) 2005-06-06 2012-10-02 Intel Corporation Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
WO2006133042A2 (en) * 2005-06-06 2006-12-14 Intel Corporation Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
US8053774B2 (en) 2005-06-06 2011-11-08 Intel Corporation Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
US9085461B2 (en) 2005-06-06 2015-07-21 Intel Corporation Method and apparatus to fabricate polymer arrays on patterned wafers using electrochemical synthesis
US10035147B2 (en) 2005-06-06 2018-07-31 Intel Corporation Wafer with gel-based biochips for electrochemical synthesis and electrical detection of polymers
US9335243B2 (en) 2006-12-11 2016-05-10 Tearlab Research, Inc. Systems and methods for collecting tear film and measuring tear film osmolarity
US8940143B2 (en) 2007-06-29 2015-01-27 Intel Corporation Gel-based bio chip for electrochemical synthesis and electrical detection of polymers
EP3260853A1 (en) * 2009-03-27 2017-12-27 ams International AG A sensor device and a method of manufacturing the same
US8470144B2 (en) 2010-12-28 2013-06-25 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US8691061B2 (en) 2010-12-28 2014-04-08 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US8691062B2 (en) 2010-12-28 2014-04-08 Japan Aviation Electronics Industry, Limited Electrode device for an electrochemical sensor chip
US9823215B2 (en) 2011-08-04 2017-11-21 Gensoric Gmbh Electrochemical sensor
WO2013017635A1 (en) * 2011-08-04 2013-02-07 Universität Rostock Electrochemical sensor
WO2013058646A1 (en) * 2011-10-19 2013-04-25 Universiti Sains Malaysia A system for identifying presence of polynucleotides of interest from a sample
TWI486586B (en) * 2013-01-16 2015-06-01 Univ Nat Chi Nan Current - type biological sensor and its making method
RU2570706C1 (en) * 2014-07-22 2015-12-10 Федеральное государственное унитарное предприятие "Государственный научный центр "Научно-исследовательский институт органических полупродуктов и красителей" (ФГУП "ГНЦ "НИОПИК") Method for qantitative determination of sodium fluorescein in substance and thereof-based medication
US11536707B2 (en) 2014-09-23 2022-12-27 Tearlab Research, Inc. Systems and methods for integration of microfluidic tear collection and lateral flow analysis of analytes of interest
US11815430B2 (en) 2019-02-22 2023-11-14 Hewlett-Packard Development Company, L.P. Nucleic acid detection
DE102021200588A1 (en) 2021-01-22 2022-07-28 Robert Bosch Gesellschaft mit beschränkter Haftung Method and control device for producing a carrier element for receiving a sample liquid, carrier element and analysis device with carrier element

Also Published As

Publication number Publication date
AU2001261145B2 (en) 2005-08-11
JP4949589B2 (en) 2012-06-13
TWI245073B (en) 2005-12-11
US7399585B2 (en) 2008-07-15
CA2407973A1 (en) 2001-11-08
CA2407973C (en) 2011-06-07
AU6114501A (en) 2001-11-12
CN100457887C (en) 2009-02-04
EP1278821A4 (en) 2005-11-09
US20020123048A1 (en) 2002-09-05
CN1440454A (en) 2003-09-03
EP1278821A1 (en) 2003-01-29
JP2003532090A (en) 2003-10-28
US8062491B1 (en) 2011-11-22

Similar Documents

Publication Publication Date Title
CA2407973C (en) Biological identification system with integrated sensor chip
AU2001261145A1 (en) Biological identification system with integrated sensor chip
Gau et al. A MEMS based amperometric detector for E. coli bacteria using self-assembled monolayers
Gooding et al. Self‐assembled monolayers into the 21st century: recent advances and applications
US20180346971A1 (en) Charge Perturbation Detection System for DNA and Other Molecules
US20190376925A1 (en) Nucleic acid sequencing device containing graphene
Zhang et al. Materials and techniques for electrochemical biosensor design and construction
US8513001B2 (en) Method and apparatus for target detection using electrode-bound viruses
WO2007114649A1 (en) Biosensor having nano wire and manufacturing method thereof
EP3147372A2 (en) Versatile and sensitive biosensor
JP5061342B2 (en) Carbon nanotube electrode and sensor using the electrode
Laschi et al. Planar electrochemical sensors for biomedical applications
Losic et al. Scanning tunneling microscopy studies of glucose oxidase on gold surfaces
Gyurcsányi et al. Chemical imaging of biological systems with the scanning electrochemical microscope
Gaspar et al. A method for the design and study of enzyme microstructures formed by means of a flow-through microdispenser
Fan et al. Disposable multiplexed electrochemical sensors based on electro-triggered selective immobilization of probes for simultaneous detection of DNA and proteins
Mosbach et al. Picodroplet-deposition of enzymes on functionalized self-assembled monolayers as a basis for miniaturized multi-sensor structures
Mohd Said Electrochemical biosensor based on microfabricated electrode arrays for life sciences applications
JP2003090815A (en) Method for electrochemically detecting gene, and nucleic acid tip
Zhang Nanoscale surface modification for enhanced biosensing
Pan Voltammetric detection of DNA hybridization using a non-competitive enzyme linked assay
Lad et al. Nanodevices for monitoring toxicological behavior of therapeutic agent
Zhu et al. An overview of Si-based biosensors
Yoon et al. Ferritin immunosensing on microfabricated electrodes based on the integration of immunoprecipitation and electrochemical signaling reactions
Lakard et al. Fabrication of a miniaturized cell using microsystem technologies for electrochemical applications

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2001261145

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2001935016

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2407973

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 018122752

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2001935016

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2001261145

Country of ref document: AU